EP2590676B1 - Virion-artige transferpartikel für selbstreplizierende rna-moleküle - Google Patents

Virion-artige transferpartikel für selbstreplizierende rna-moleküle Download PDF

Info

Publication number
EP2590676B1
EP2590676B1 EP11741348.4A EP11741348A EP2590676B1 EP 2590676 B1 EP2590676 B1 EP 2590676B1 EP 11741348 A EP11741348 A EP 11741348A EP 2590676 B1 EP2590676 B1 EP 2590676B1
Authority
EP
European Patent Office
Prior art keywords
rna
virus
particle
liposome
liposomes
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Revoked
Application number
EP11741348.4A
Other languages
English (en)
French (fr)
Other versions
EP2590676A2 (de
Inventor
Andrew Geall
Christian Mandl
Derek O'hagan
Manmohan Singh
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
GlaxoSmithKline Biologicals SA
Original Assignee
GlaxoSmithKline Biologicals SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=44629863&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP2590676(B1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to SI201130994A priority Critical patent/SI2590676T1/sl
Priority to RS20160879A priority patent/RS55260B1/sr
Priority to EP22200367.5A priority patent/EP4180057A1/de
Priority to EP16184271.1A priority patent/EP3115061A1/de
Priority to EP21204155.2A priority patent/EP4005592B1/de
Application filed by GlaxoSmithKline Biologicals SA filed Critical GlaxoSmithKline Biologicals SA
Publication of EP2590676A2 publication Critical patent/EP2590676A2/de
Publication of EP2590676B1 publication Critical patent/EP2590676B1/de
Application granted granted Critical
Priority to HRP20161352TT priority patent/HRP20161352T1/hr
Revoked legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/155Paramyxoviridae, e.g. parainfluenza virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/245Herpetoviridae, e.g. herpes simplex virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55566Emulsions, e.g. Freund's adjuvant, MF59
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16111Cytomegalovirus, e.g. human herpesvirus 5
    • C12N2710/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18522New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18534Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/36011Togaviridae
    • C12N2770/36111Alphavirus, e.g. Sindbis virus, VEE, EEE, WEE, Semliki
    • C12N2770/36141Use of virus, viral particle or viral elements as a vector
    • C12N2770/36143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention is in the field of non-viral delivery of self-replicating RNAs for immunisation.
  • nucleic acids for immunising animals has been a goal for several years.
  • Various approaches have been tested, including the use of DNA or RNA, of viral or non-viral delivery vehicles (or even no delivery vehicle, in a "naked” vaccine), of replicating or non-replicating vectors, or of viral or non-viral vectors.
  • Zhou et al. Human Gene Therapy, 1999, vol. 10, no. 16, p. 2719-2724
  • Saeki et al. Human Gene Therapy, 1997, vol. 8, no. 17, p. 2133-2141
  • HVJ-liposomes which comprise a protein capsid.
  • Levine et al. (Nature Immunology, 2004, vol. 5, no. 5, p.
  • 460-464 reports packaging of replicon RNA into virions.
  • Mockey et al. Cancer Gene Therapy, 2007, vol. 14, no. 9, p. 802-814
  • Martinon et al. European Journal of Immunology, 1993, vol. 23, no. 7, p. 1719-1722
  • Cannon et al. (DNA and Cell Biology, 2002, vol. 21, no. 12, p. 953-961 ) reports data in which RNA was packaged into virions, used in naked form, or adsorbed to gold particles.
  • nucleic acid vaccines There remains a need for further and improved nucleic acid vaccines and, in particular, for improved ways of delivering nucleic acid vaccines.
  • nucleic acid immunisation is achieved by delivering a self-replicating RNA encapsulated within a small particle.
  • the RNA encodes an immunogen of interest, and the particle may deliver this RNA by mimicking the delivery function of a natural virus.
  • the invention provides a non-virion particle that does not comprise a protein capsid, for in vivo delivery of RNA to a vertebrate cell, wherein the particle is a liposome and comprises a delivery material encapsulating a self-replicating RNA molecule which encodes an immunogen, wherein the immunogen is a surface polypeptide and can elicit an immune response in vivo against a bacterium, a virus, a fungus or a parasite, and the RNA includes no modified nucleotides, and optionally includes a 5' cap.
  • the particles are useful as components in pharmaceutical compositions for immunising subjects against various diseases.
  • Particles of the invention are non-virion particles i. e. they are not a virion.
  • the particle does not comprise a protein capsid.
  • the invention does not require a packaging cell line, thus permitting easier up-scaling for commercial production and minimising the risk that dangerous infectious viruses will inadvertently be produced.
  • particles of the invention are formed from a delivery material.
  • Various materials are suitable for forming particles which can deliver RNA to a vertebrate cell in vivo. Delivery materials of particular interest are amphiphilic lipids which can form liposomes. Where delivery is by liposome, RNA should be encapsulated.
  • a particle of the invention comprises a liposome encapsulating a self-replicating RNA molecule which encodes an immunogen, as defined in the claims.
  • the particles preferably are substantially spherical.
  • RNA is encapsulated within the particles (the particle is a liposome).
  • the particle is a liposome.
  • RNA inside the particles is (as in a natural virus) separated from any external medium by the delivery material, and encapsulation has been found to protect RNA from RNase digestion.
  • Encapsulation can take various forms. For example, in some embodiments (as in a unilamellar liposome) the delivery material forms a outer layer around an aqueous RNA-containing core.
  • the particles can include some external RNA (e.g. on the surface of the particles), but at least half of the RNA (and ideally all of it) is encapsulated. Encapsulation within liposomes is distinct from, for instance, the lipid/RNA complexes disclosed in reference 1.
  • RNA-containing aqueous core can have an anionic, cationic or zwitterionic hydrophilic head group.
  • anionic phospholipids dates back to the 1960s, and cationic liposome-forming lipids have been studied since the 1990s.
  • Some phospholipids are anionic whereas other are zwitterionic and others are cationic.
  • Suitable classes of phospholipid include, but are not limited to, phosphatidylethanolamines, phosphatidylcholines, phosphatidylserines, and phosphatidyl-glycerols, and some useful phospholipids are listed in Table 1.
  • Useful cationic lipids include, but are not limited to, dioleoyl trimethylammonium propane (DOTAP), 1,2-distearyloxy-N,N-dimethyl-3-aminopropane (DSDMA), 1,2-dioleyloxy-N,Ndimethyl-3-aminopropane (DODMA), 1,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane (DLinDMA), 1,2-dilinolenyloxy-N,N-dimethyl-3-aminopropane (DLenDMA).
  • Zwitterionic lipids include, but are not limited to, acyl zwitterionic lipids and ether zwitterionic lipids.
  • lipids can be saturated or unsaturated.
  • the use of at least one unsaturated lipid for preparing liposomes is preferred. If an unsaturated lipid has two tails, both tails can be unsaturated, or it can have one saturated tail and one unsaturated tail.
  • Liposomal particles of the invention can be formed from a single lipid or from a mixture of lipids.
  • a mixture may comprise (i) a mixture of anionic lipids (ii) a mixture of cationic lipids (iii) a mixture of zwitterionic lipids (iv) a mixture of anionic lipids and cationic lipids (v) a mixture of anionic lipids and zwitterionic lipids (vi) a mixture of zwitterionic lipids and cationic lipids or (vii) a mixture of anionic lipids, cationic lipids and zwitterionic lipids.
  • a mixture may comprise both saturated and unsaturated lipids.
  • a mixture may comprise DSPC (zwitterionic, saturated), DlinDMA (cationic, unsaturated), and/or DMG (anionic, saturated).
  • DSPC zwitterionic, saturated
  • DlinDMA cationic, unsaturated
  • DMG anionic, saturated
  • the hydrophilic portion of a lipid can be PEGylated ( i.e. modified by covalent attachment of a polyethylene glycol). This modification can increase stability and prevent non-specific adsorption of the liposomes.
  • lipids can be conjugated to PEG using techniques such as those disclosed in reference 2 and 3.
  • Various lengths of PEG can be used e.g. between 0.5-8kDa.
  • a mixture of DSPC, DlinDMA, PEG-DMG and cholesterol is used in the examples.
  • Liposomal particles are usually divided into three groups: multilamellar vesicles (MLV); small unilamellar vesicles (SUV); and large unilamellar vesicles (LUV).
  • MLVs have multiple bilayers in each vesicle, forming several separate aqueous compartments.
  • SUVs and LUVs have a single bilayer encapsulating an aqueous core; SUVs typically have a diameter ⁇ 50nm, and LUVs have a diameter >50nm.
  • Liposomal particles of the invention are ideally LUVs with a diameter in the range of 50-220nm.
  • compositions comprising a population of LUVs with different diameters: (i) at least 80% by number should have diameters in the range of 20-220nm, (ii) the average diameter (Zav, by intensity) of the population is ideally in the range of 40-200nm, and/or (iii) the diameters should have a polydispersity index ⁇ 0.2.
  • the liposome/RNA complexes of reference 1 are expected to have a diameter in the range of 600-800nm and to have a high polydispersity.
  • Particles of the invention include a self-replicating RNA molecule which (unlike siRNA) encodes an immunogen. After in vivo administration of the particles, RNA is released from the particles and is translated inside a cell to provide the immunogen in situ.
  • RNA is released from the particles and is translated inside a cell to provide the immunogen in situ.
  • RNA in particles of the invention is self-replicating.
  • a self-replicating RNA molecule can, when delivered to a vertebrate cell even without any proteins, lead to the production of multiple daughter RNAs by transcription from itself (via an antisense copy which it generates from itself).
  • a self-replicating RNA molecule is thus typically a +-strand molecule which can be directly translated after delivery to a cell, and this translation provides a RNA-dependent RNA polymerase which then produces both antisense and sense transcripts from the delivered RNA.
  • the delivered RNA leads to the production of multiple daughter RNAs.
  • RNAs may be translated themselves to provide in situ expression of an encoded immunogen, or may be transcribed to provide further transcripts with the same sense as the delivered RNA which are translated to provide in situ expression of the immunogen.
  • the overall results of this sequence of transcriptions is a huge amplification in the number of the introduced replicon RNAs and so the encoded immunogen becomes a major polypeptide product of the cells.
  • One suitable system for achieving self-replication in this manner is to use an alphavirus-based replicon.
  • These replicons are +-stranded RNAs which lead to translation of a replicase (or replicase-transcriptase) after delivery to a cell.
  • the replicase is translated as a polyprotein which auto-cleaves to provide a replication complex which creates genomic --strand copies of the +-strand delivered RNA.
  • These --strand transcripts can themselves be transcribed to give further copies of the +-stranded parent RNA and also to give a subgenomic transcript which encodes the immunogen. Translation of the subgenomic transcript thus leads to in situ expression of the immunogen by the infected cell.
  • Suitable alphavirus replicons can use a replicase from a Sindbis virus, a Semliki forest virus, an eastern equine encephalitis virus, a Venezuelan equine encephalitis virus, etc.
  • Mutant or wild-type virus sequences can be used e.g. the attenuated TC83 mutant of VEEV has been used in replicons [14].
  • a preferred self-replicating RNA molecule thus encodes (i) a RNA-dependent RNA polymerase which can transcribe RNA from the self-replicating RNA molecule and (ii) an immunogen.
  • the polymerase can be an alphavirus replicase e.g. comprising one or more of alphavirus proteins nsP1, nsP2, nsP3 and nsP4.
  • the self-replicating RNA molecules of the invention do not encode alphavirus structural proteins.
  • a preferred self-replicating RNA can lead to the production of genomic RNA copies of itself in a cell, but not to the production of RNA-containing virions.
  • the inability to produce these virions means that, unlike a wild-type alphavirus, the self-replicating RNA molecule cannot perpetuate itself in infectious form.
  • alphavirus structural proteins which are necessary for perpetuation in wild-type viruses are absent from self-replicating RNAs of the invention and their place is taken by gene(s) encoding the immunogen of interest, such that the subgenomic transcript encodes the immunogen rather than the structural alphavirus virion proteins.
  • RNA molecule useful with the invention may have two open reading frames.
  • the first (5') open reading frame encodes a replicase; the second (3') open reading frame encodes an immunogen.
  • the RNA may have additional (e.g. downstream) open reading frames e.g. to encode further immunogens (see below) or to encode accessory polypeptides.
  • a preferred self-replicating RNA molecule has a 5' cap (e.g. a 7-methylguanosine). This cap can enhance in vivo translation of the RNA.
  • the 5' sequence of the self-replicating RNA molecule must be selected to ensure compatibility with the encoded replicase.
  • a self-replicating RNA molecule may have a 3' poly-A tail. It may also include a poly-A polymerase recognition sequence (e.g. AAUAAA) near its 3' end.
  • AAUAAA poly-A polymerase recognition sequence
  • Self-replicating RNA molecules can have various lengths but they are typically 5000-25000 nucleotides long e.g. 8000-15000 nucleotides, or 9000-12000 nucleotides. Thus the RNA is longer than seen in siRNA delivery.
  • RNA molecules will typically be single-stranded.
  • Single-stranded RNAs can generally initiate an adjuvant effect by binding to TLR7, TLR8, RNA helicases and/or PKR.
  • RNA delivered in double-stranded form can bind to TLR3, and this receptor can also be triggered by dsRNA which is formed either during replication of a single-stranded RNA or within the secondary structure of a single-stranded RNA.
  • the self-replicating RNA can conveniently be prepared by in vitro transcription (IVT).
  • IVT can use a (cDNA) template created and propagated in plasmid form in bacteria, or created synthetically (for example by gene synthesis and/or polymerase chain-reaction (PCR) engineering methods).
  • a DNA-dependent RNA polymerase such as the bacteriophage T7, T3 or SP6 RNA polymerases
  • Appropriate capping and poly-A addition reactions can be used as required (although the replicon's poly-A is usually encoded within the DNA template).
  • RNA polymerases can have stringent requirements for the transcribed 5' nucleotide(s) and in some embodiments these requirements must be matched with the requirements of the encoded replicase, to ensure that the IVT-transcribed RNA can function efficiently as a substrate for its self-encoded replicase.
  • the RNA includes no modified nucleotides i. e. all of the nucleotides in the RNA are standard A, C, G and U ribonucleotides (except for any 5' cap structure, which may include a 7'-methylguanosine).
  • the RNA may include a 5' cap comprising a 7'-methylguanosine, and the first 1, 2 or 3 5' ribonucleotides may be methylated at the 2' position of the ribose.
  • a RNA used with the invention ideally includes only phosphodiester linkages between nucleosides, but in some embodiments it can contain phosphoramidate, phosphorothioate, and/or methylphosphonate linkages.
  • RNA per particle can vary, and the number of individual self-replicating RNA molecules per particle can depend on the characteristics of the particle being used.
  • a particle may include from 1-500 RNA molecules.
  • the number of RNA molecules is typically ⁇ 50 per liposome e.g. ⁇ 20, ⁇ 10, ⁇ 5, or 1-4.
  • the number of RNA molecules will depend on the particle diameter but may be ⁇ 50 per particle (e.g. ⁇ 20, ⁇ 10, ⁇ 5, or 1-4) or from 50-200 per particle.
  • a particle includes fewer than 10 different species of RNA e.g. 5, 4, 3, or 2 different species; most preferably, a particle includes a single RNA species i. e. all RNA molecules in the particle have the same sequence and same length.
  • RNA molecules used with the invention encode a polypeptide immunogen. After administration of the particles the immunogen is translated in vivo and can elicit an immune response in the recipient against a bacterium, a virus, a fungus or a parasite (and, in some embodiments, against an allergen; and in other embodiments, against a tumor antigen).
  • the immune response may comprise an antibody response (usually including IgG) and/or a cell-mediated immune response.
  • the polypeptide immunogen will typically elicit an immune response which recognises the corresponding bacterial, viral, fungal or parasite (or allergen or tumour) polypeptide, but in some embodiments the polypeptide may act as a mimotope to elicit an immune response which recognises a bacterial, viral, fungal or parasite saccharide.
  • the immunogen is a surface polypeptide e.g. an adhesin, a hemagglutinin, an envelope glycoprotein, a spike glycoprotein, etc.
  • Self-replicating RNA molecules can encode a single polypeptide immunogen or multiple polypeptides. Multiple immunogens can be presented as a single polypeptide immunogen (fusion polypeptide) or as separate polypeptides. If immunogens are expressed as separate polypeptides then one or more of these may be provided with an upstream IRES or an additional viral promoter element. Alternatively, multiple immunogens may be expressed from a polyprotein that encodes individual immunogens fused to a short autocatalytic protease (e.g. foot-and-mouth disease virus 2A protein), or as inteins.
  • a short autocatalytic protease e.g. foot-and-mouth disease virus 2A protein
  • the RNA encodes an immunogen.
  • the invention does not encompass RNA which encodes a firefly luciferase or which encodes a fusion protein of E.coli ⁇ -galactosidase or which encodes a green fluorescent protein (GFP).
  • GFP green fluorescent protein
  • the RNA is not total mouse thymus RNA.
  • the immunogen elicits an immune response against a virus which infects fish, such as: infectious salmon anemia virus (ISAV), salmon pancreatic disease virus (SPDV), infectious pancreatic necrosis virus (IPNV), channel catfish virus (CCV), fish lymphocystis disease virus (FLDV), infectious hematopoietic necrosis virus (IHNV), koi herpesvirus, salmon picorna-like virus (also known as picorna-like virus of atlantic salmon), landlocked salmon virus (LSV), atlantic salmon rotavirus (ASR), trout strawberry disease virus (TSD), coho salmon tumor virus (CSTV), or viral hemorrhagic septicemia virus (VHSV).
  • infectious salmon anemia virus ISAV
  • SPDV salmon pancreatic disease virus
  • IPNV infectious pancreatic necrosis virus
  • CCV channel catfish virus
  • FLDV fish lymphocystis disease virus
  • IHNV infectious hematopoietic necrosis virus
  • Fungal immunogens may be derived from Dermatophytres, including: Epidermophyton floccusum, Microsporum audouini, Microsporum canis, Microsporum distortum, Microsporum equinum, Microsporum gypsum, Microsporum nanum , Trichophyton concentricum, Trichophyton equinum, Trichophyton gallinae, Trichophyton gypseum, Trichophyton naegnini, Trichophyton mentagrophytes , Trichophyton quinckeanum, Trichophyton rubrum , Trichophyton schoenleini, Trichophyton tonsurans , Trichophyton verrucosum , T.
  • Dermatophytres including: Epidermophyton floccusum, Microsporum audouini, Microsporum canis, Microsporum distortum, Microsporum equinum, Microsporum gypsum, Microsporum nanum
  • the immunogen elicits an immune response against a parasite from the Plasmodium genus, such as P.falciparum, P.vivax, P.malariae or P.ovale.
  • the invention may be used for immunising against malaria.
  • the immunogen elicits an immune response against a parasite from the Caligidae family, particularly those from the Lepeophtheirus and Caligus genera e.g. sea lice such as Lepeophtheirus salmonis or Caligus rogercresseyi.
  • the immunogen elicits an immune response against: pollen allergens (tree-, herb, weed-, and grass pollen allergens); insect or arachnid allergens (inhalant, saliva and venom allergens, e.g. mite allergens, cockroach and midges allergens, hymenopthera venom allergens); animal hair and dandruff allergens (from e.g. dog, cat, horse, rat, mouse, etc.); and food allergens (e.g. a gliadin).
  • pollen allergens tree-, herb, weed-, and grass pollen allergens
  • insect or arachnid allergens inhalant, saliva and venom allergens, e.g. mite allergens, cockroach and midges allergens, hymenopthera venom allergens
  • animal hair and dandruff allergens from e.g. dog, cat, horse
  • Important pollen allergens from trees, grasses and herbs are such originating from the taxonomic orders of Fagales, Oleales, Pinales and platanaceae including, but not limited to, birch (Betula), alder (Alnus), hazel (Corylus), hornbeam (Carpinus) and olive (Olea), cedar (Cryptomeria and Juniperus), plane tree (Platanus), the order of Poales including grasses of the genera Lolium, Phleum, Poa, Cynodon, Dactylis, Holcus, Phalaris, Secale, and Sorghum, the orders of Asterales and Urticales including herbs of the genera Ambrosia, Artemisia, and Parietaria.
  • venom allergens including such originating from stinging or biting insects such as those from the taxonomic order of Hymenoptera including bees ( Apidae ) , wasps ( Vespidea ) , and ants ( Formicoidae ) .
  • the immunogen is a tumor antigen selected from: (a) cancer-testis antigens such as NY-ESO-1, SSX2, SCP1 as well as RAGE, BAGE, GAGE and MAGE family polypeptides, for example, GAGE-1, GAGE-2, MAGE-1, MAGE-2, MAGE-3, MAGE-4, MAGE-5, MAGE-6, and MAGE-12 (which can be used, for example, to address melanoma, lung, head and neck, NSCLC, breast, gastrointestinal, and bladder tumors; (b) mutated antigens, for example, p53 (associated with various solid tumors, e.g., colorectal, lung, head and neck cancer), p21/Ras (associated with, e.g., melanoma, pancreatic cancer and colorectal cancer), CDK4 (associated with, e.g., melanoma), MUM1 (associated with, e.g., melanoma), caspase-8
  • tumor immunogens include, but are not limited to, p 15, Hom/Mel-40, H-Ras, E2A-PRL, H4-RET, IGH-IGK, MYL-RAR, Epstein Barr virus antigens, EBNA, human papillomavirus (HPV) antigens, including E6 and E7, hepatitis B and C virus antigens, human T-cell lymphotropic virus antigens, TSP-180, p185erbB2, p180erbB-3, c-met, mn-23H1, TAG-72-4, CA 19-9, CA 72-4, CAM 17.1, NuMa, K-ras, p16, TAGE, PSCA, CT7, 43-9F, 5T4, 791 Tgp72, beta-HCG, BCA225, BTAA, CA 125, CA 15-3 (CA 27.29 ⁇ BCAA), CA 195, CA 242, CA-50, CAM43, CD68 ⁇ KP1, CO
  • Particles of the invention are useful as components in pharmaceutical compositions for immunising subjects against various diseases. These compositions will typically include a pharmaceutically acceptable carrier in addition to the particles. A thorough discussion of pharmaceutically acceptable carriers is available in reference 35.
  • a pharmaceutical composition of the invention may include one or more small molecule immunopotentiators.
  • the composition may include a TLR2 agonist (e.g. Pam3CSK4), a TLR4 agonist (e.g. an aminoalkyl glucosaminide phosphate, such as E6020), a TLR7 agonist (e.g. imiquimod), a TLR8 agonist (e.g. resiquimod) and/or a TLR9 agonist (e.g. IC31).
  • a TLR2 agonist e.g. Pam3CSK4
  • TLR4 agonist e.g. an aminoalkyl glucosaminide phosphate, such as E6020
  • TLR7 agonist e.g. imiquimod
  • a TLR8 agonist e.g. resiquimod
  • TLR9 agonist e.g. IC31
  • Any such agonist ideally has a molecular weight of ⁇ 2000D
  • compositions of the invention may include the particles in plain water (e.g. w.f.i.) or in a buffer e.g. a phosphate buffer, a Tris buffer, a borate buffer, a succinate buffer, a histidine buffer, or a citrate buffer.
  • Buffer salts will typically be included in the 5-20mM range.
  • compositions of the invention may have a pH between 5.0 and 9.5 e.g. between 6.0 and 8.0.
  • compositions of the invention may include sodium salts (e.g. sodium chloride) to give tonicity.
  • sodium salts e.g. sodium chloride
  • a concentration of 10 ⁇ 2 mg/ml NaCl is typical e.g. about 9 mg/ml.
  • compositions of the invention may include metal ion chelators. These can prolong RNA stability by removing ions which can accelerate phosphodiester hydrolysis.
  • a composition may include one or more of EDTA, EGTA, BAPTA, pentetic acid, etc..
  • chelators are typically present at between 10-500 ⁇ M e.g. 0.1mM.
  • a citrate salt, such as sodium citrate, can also act as a chelator, while advantageously also providing buffering activity.
  • compositions of the invention may have an osmolality of between 200 mOsm/kg and 400 mOsm/kg, e.g. between 240-360 mOsm/kg, or between 290-310 mOsm/kg.
  • compositions of the invention may include one or more preservatives, such as thiomersal or 2-phenoxyethanol.
  • preservatives such as thiomersal or 2-phenoxyethanol.
  • Mercury-free compositions are preferred, and preservative-free vaccines can be prepared.
  • compositions of the invention are preferably sterile.
  • compositions of the invention are preferably non-pyrogenic e.g. containing ⁇ 1 EU (endotoxin unit, a standard measure) per dose, and preferably ⁇ 0.1 EU per dose.
  • ⁇ 1 EU endotoxin unit, a standard measure
  • compositions of the invention are preferably gluten free.
  • compositions of the invention may be prepared in unit dose form.
  • a unit dose may have a volume of between 0.1-1.0ml e.g. about 0.5ml.
  • compositions may be prepared as injectables, either as solutions or suspensions.
  • the composition may be prepared for pulmonary administration e.g. by an inhaler, using a fine spray.
  • the composition may be prepared for nasal, aural or ocular administration e.g. as spray or drops. Injectables for intramuscular administration are typical.
  • compositions comprise an immunologically effective amount of particles, as well as any other components, as needed.
  • 'immunologically effective amount' it is meant that the administration of that amount to an individual, either in a single dose or as part of a series, is effective for treatment or prevention. This amount varies depending upon the health and physical condition of the individual to be treated, age, the taxonomic group of individual to be treated (e.g. non-human primate, primate, etc.), the capacity of the individual's immune system to synthesise antibodies, the degree of protection desired, the formulation of the vaccine, the treating doctor's assessment of the medical situation, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials.
  • compositions of the invention will generally be expressed in terms of the amount of RNA per dose.
  • a preferred dose has ⁇ 100 ⁇ g RNA (e.g. from 10-100 ⁇ g, such as about 10 ⁇ g, 25 ⁇ g, 50 ⁇ g, 75 ⁇ g or 100 ⁇ g), but expression can be seen at much lower levels e.g. ⁇ 1 ⁇ g/dose, ⁇ 100ng/dose, ⁇ 10ng/dose, ⁇ 1ng/dose, etc
  • the invention also provides a delivery device (e.g. syringe, nebuliser, sprayer, inhaler, dermal patch, etc.) containing a pharmaceutical composition of the invention.
  • a delivery device e.g. syringe, nebuliser, sprayer, inhaler, dermal patch, etc.
  • This device can be used to administer the composition to a vertebrate subject.
  • Particles of the invention do not include ribosomes.
  • particles and pharmaceutical compositions of the invention are for in vivo use for eliciting an immune response against an immunogen of interest.
  • the invention provides a particle or pharmaceutical composition of the invention for use in a method for raising an immune response in a vertebrate comprising the step of administering an effective amount of a particle or pharmaceutical composition of the invention.
  • the immune response is protective and preferably involves antibodies and/or cell-mediated immunity.
  • the method may raise a booster response.
  • the vertebrate By raising an immune response in the vertebrate by these uses and methods, the vertebrate can be protected against various diseases and/or infections e.g. against bacterial and/or viral diseases as discussed above.
  • the particles and compositions are immunogenic, and are more preferably vaccine compositions.
  • Vaccines according to the invention may either be prophylactic (i.e. to prevent infection) or therapeutic (i. e. to treat infection), but will typically be prophylactic.
  • the vertebrate is preferably a mammal, such as a human or a large veterinary mammal (e.g. horses, cattle, deer, goats, pigs).
  • the human is preferably a child (e.g. a toddler or infant) or a teenager; where the vaccine is for therapeutic use, the human is preferably a teenager or an adult.
  • a vaccine intended for children may also be administered to adults e.g. to assess safety, dosage, immunogenicity, etc.
  • Vaccines prepared according to the invention may be used to treat both children and adults.
  • a human patient may be less than 1 year old, less than 5 years old, 1-5 years old, 5-15 years old, 15-55 years old, or at least 55 years old.
  • Preferred patients for receiving the vaccines are the elderly (e.g. ⁇ 50 years old, ⁇ 60 years old, and preferably ⁇ 65 years), the young ( e.g. ⁇ 5 years old), hospitalised patients, healthcare workers, armed service and military personnel, pregnant women, the chronically ill, or immunodeficient patients.
  • the vaccines are not suitable solely for these groups, however, and may be used more generally in a population.
  • compositions of the invention will generally be administered directly to a patient.
  • Direct delivery may be accomplished by parenteral injection (e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, intradermally, or to the interstitial space of a tissue; unlike reference 1, intraglossal injection is not typically used with the present invention).
  • Alternative delivery routes include rectal, oral (e.g. tablet, spray), buccal, sublingual, vaginal, topical, transdermal or transcutaneous, intranasal, ocular, aural, pulmonary or other mucosal administration.
  • Intradermal and intramuscular administration are two preferred routes. Injection may be via a needle (e.g. a hypodermic needle), but needle-free injection may alternatively be used.
  • a typical intramuscular dose is 0.5 ml.
  • the invention may be used to elicit systemic and/or mucosal immunity, preferably to elicit an enhanced systemic and/or mucosal immunity.
  • Dosage can be by a single dose schedule or a multiple dose schedule. Multiple doses may be used in a primary immunisation schedule and/or in a booster immunisation schedule. In a multiple dose schedule the various doses may be given by the same or different routes e.g. a parenteral prime and mucosal boost, a mucosal prime and parenteral boost, etc. Multiple doses will typically be administered at least 1 week apart (e.g. about 2 weeks, about 3 weeks, about 4 weeks, about 6 weeks, about 8 weeks, about 10 weeks, about 12 weeks, about 16 weeks, etc.). In one embodiment, multiple doses may be administered approximately 6 weeks, 10 weeks and 14 weeks after birth, e.g.
  • two primary doses are administered about two months apart, e.g. about 7, 8 or 9 weeks apart, followed by one or more booster doses about 6 months to 1 year after the second primary dose, e.g. about 6, 8, 10 or 12 months after the second primary dose.
  • three primary doses are administered about two months apart, e.g. about 7, 8 or 9 weeks apart, followed by one or more booster doses about 6 months to 1 year after the third primary dose, e.g. about 6, 8, 10, or 12 months after the third primary dose.
  • composition comprising X may consist exclusively of X or may include something additional e.g. X + Y.
  • TLR3 is the Toll-like receptor 3. It is a single membrane-spanning receptor which plays a key role in the innate immune system.
  • Known TLR3 agonists include poly(I:C).
  • TLR3 is the approved HGNC name for the gene encoding this receptor, and its unique HGNC ID is HGNC: 11849.
  • the RefSeq sequence for the human TLR3 gene is GI:2459625.
  • TLR7 is the Toll-like receptor 7. It is a single membrane-spanning receptor which plays a key role in the innate immune system.
  • Known TLR7 agonists include e.g. imiquimod.
  • TLR7 is the approved HGNC name for the gene encoding this receptor, and its unique HGNC ID is HGNC:15631.
  • the RefSeq sequence for the human TLR7 gene is GI:67944638.
  • TLR8 is the Toll-like receptor 8. It is a single membrane-spanning receptor which plays a key role in the innate immune system.
  • Known TLR8 agonists include e.g. resiquimod.
  • TLR8 is the approved HGNC name for the gene encoding this receptor, and its unique HGNC ID is HGNC:15632.
  • the RefSeq sequence for the human TLR8 gene is GI:20302165.
  • RLR-1 The RIG-I-like receptor (“RLR”) family includes various RNA helicases which play key roles in the innate immune system[43].
  • RLR-1 also known as RIG-I or retinoic acid inducible gene I
  • RLR-1 helicase has two caspase recruitment domains near its N-terminus.
  • the approved HGNC name for the gene encoding the RLR-1 helicase is "DDX58" (for DEAD (Asp-Glu-Ala-Asp) box polypeptide 58) and the unique HGNC ID is HGNC:19102.
  • the RefSeq sequence for the human RLR-1 gene is GI:77732514.
  • RLR-2 (also known as MDA5 or melanoma differentiation-associated gene 5) also has two caspase recruitment domains near its N-terminus.
  • the approved HGNC name for the gene encoding the RLR-2 helicase is "IFIH1" (for interferon induced with helicase C domain 1) and the unique HGNC ID is HGNC: 18873.
  • the RefSeq sequence for the human RLR-2 gene is GI: 27886567.
  • RLR-3 (also known as LGP2 or laboratory of genetics and physiology 2) has no caspase recruitment domains.
  • the approved HGNC name for the gene encoding the RLR-3 helicase is "DHX58" (for DEXH (Asp-Glu-X-His) box polypeptide 58) and the unique HGNC ID is HGNC:29517.
  • the RefSeq sequence for the human RLR-3 gene is GI:149408121.
  • PKR is a double-stranded RNA-dependent protein kinase. It plays a key role in the innate immune system.
  • EIF2AK2 for eukaryotic translation initiation factor 2-alpha kinase 2
  • HGNC HGNC:9437
  • the RefSeq sequence for the human PKR gene is GI:208431825.
  • replicons are used below. In general these are based on a hybrid alphavirus genome with non-structural proteins from venezuelan equine encephalitis virus (VEEV), a packaging signal from Sindbis virus, and a 3' UTR from Sindbis virus or a VEEV mutant.
  • VEEV venezuelan equine encephalitis virus
  • Sindbis virus Sindbis virus
  • the replicon is about 10kb long and has a poly-A tail.
  • Plasmid DNA encoding alphavirus replicons (named: pT7-mVEEV-FL.RSVF or A317; pT7-mVEEV-SEAP or A306; pSP6-VCR-GFP or A50) served as a template for synthesis of RNA in vitro.
  • the replicons contain the alphavirus genetic elements required for RNA replication but lack those encoding gene products necessary for particle assembly; the structural proteins are instead replaced by a protein of interest (either a reporter, such as SEAP or GFP, or an immunogen, such as full-length RSV F protein) and so the replicons are incapable of inducing the generation of infectious particles.
  • a bacteriophage (T7 or SP6) promoter upstream of the alphavirus cDNA facilitates the synthesis of the replicon RNA in vitro and a hepatitis delta virus (HDV) ribozyme immediately downstream of the poly(A)-tail generates the correct 3'-end through its self-cleaving activity.
  • HDV hepatitis delta virus
  • run-off transcripts were synthesized in vitro using T7 or SP6 bacteriophage derived DNA-dependent RNA polymerase. Transcriptions were performed for 2 hours at 37°C in the presence of 7.5 mM (T7 RNA polymerase) or 5 mM (SP6 RNA polymerase) of each of the nucleoside triphosphates (ATP, CTP, GTP and UTP) following the instructions provided by the manufacturer (Ambion). Following transcription the template DNA was digested with TURBO DNase (Ambion).
  • RNA was precipitated with LiCl and reconstituted in nuclease-free water.
  • Uncapped RNA was capped post-transcriptionally with Vaccinia Capping Enzyme (VCE) using the ScriptCap m7G Capping System (Epicentre Biotechnologies) as outlined in the user manual; replicons capped in this way are given the "v" prefix e.g. vA317 is the A317 replicon capped by VCE.
  • Post-transcriptionally capped RNA was precipitated with LiCl and reconstituted in nuclease-free water. The concentration of the RNA samples was determined by measuring OD 260nm . Integrity of the in vitro transcripts was confirmed by denaturing agarose gel electrophoresis.
  • Microparticles were made using 500mg of PLG RG503 (50:50 lactide/glycolide molar ratio, MW ⁇ 30kDa) and 20mg DOTAP using an Omni Macro Homogenizer. The particle suspension was shaken at 150rpm overnight and then filtered through a 40 ⁇ m sterile filter for storage at 2-8 °C. Self-replicating RNA was adsorbed to the particles. To prepare 1 mL of PLG/RNA suspension the required volume of PLG particle suspension was added to a vial and nuclease-free water was added to bring the volume to 900 ⁇ L. 100 ⁇ L RNA (10 ⁇ g/mL) was added dropwise to the PLG suspension, with constant shaking.
  • PLG/RNA was incubated at room temperature for 30 min. For 1 mL of reconstituted suspension, 45mg mannitol, 15mg sucrose and 250-500 ⁇ g of PVA were added. The vials were frozen at -80°C and lyophilized.
  • RNA adsorption 100 ⁇ L particle suspension was centrifuged at 10,000 rpm for 5 min and supernatant was collected. PLG/RNA was reconstituted using 1mL nuclease-free water. To 100 ⁇ L particle suspension (1 ⁇ g RNA), 1mg heparin sulfate was added. The mixture was vortexed and allowed to sit at room temperature for 30 min for RNA desorption. Particle suspension was centrifuged and supernatant was collected.
  • RNAse stability 100 ⁇ L particle suspension was incubated with 6.4mAU of RNase A at room temperature for 30 min. RNAse was inactivated with 0.126mAU of Proteinase K at 55°C for 10 min. 1mg of heparin sulfate was added to desorb the RNA followed by centrifugation. The supernatant samples containing RNA were mixed with formaldehyde load dye, heated at 65°C for 10 min and analyzed using a 1% denaturing gel (460ng RNA loaded per lane).
  • RNA was encapsulated in liposomes made by the method of references 7 and 44.
  • the liposomes were made of 10% DSPC (zwitterionic), 40% DlinDMA (cationic), 48% cholesterol and 2% PEG-conjugated DMG (2kDa PEG). These proportions refer to the % moles in the total liposome.
  • DlinDMA (1,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane) was synthesized using the procedure of reference 2.
  • DSPC (1,2-Diastearoyl-sn-glycero-3-phosphocholine) was purchased from Genzyme. Cholesterol was obtained from Sigma-Aldrich.
  • PEG-conjugated DMG (1,2-dimyristoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol), ammonium salt), DOTAP (1,2-dioleoyl-3-trimethylammonium-propane, chloride salt) and DC-chol (3 ⁇ -[N-(N',N'-dimethylaminoethane)-carbamoyl]cholesterol hydrochloride) were from Avanti Polar Lipids.
  • lipids were dissolved in ethanol (2ml), a RNA replicon was dissolved in buffer (2ml, 100mM sodium citrate, pH 6) and these were mixed with 2ml of buffer followed by 1 hour of equilibration. The mixture was diluted with 6ml buffer then filtered. The resulting product contained liposomes, with ⁇ 95% encapsulation efficiency.
  • fresh lipid stock solutions were prepared in ethanol.
  • 37 mg of DlinDMA, 11.8 mg of DSPC, 27.8 mg of cholesterol and 8.07 mg of PEG-DMG were weighed and dissolved in 7.55 mL of ethanol.
  • the freshly prepared lipid stock solution was gently rocked at 37°C for about 15 min to form a homogenous mixture.
  • 755 ⁇ L of the stock was added to 1.245 mL ethanol to make a working lipid stock solution of 2 mL. This amount of lipids was used to form liposomes with 250 ⁇ g RNA.
  • RNA working solution was also prepared from a stock solution of ⁇ 1 ⁇ g/ ⁇ L in 100 mM citrate buffer (pH 6). Three 20 mL glass vials (with stir bars) were rinsed with RNase Away solution (Molecular BioProducts) and washed with plenty of MilliQ water before use to decontaminate the vials of RNases. One of the vials was used for the RNA working solution and the others for collecting the lipid and RNA mixes (as described later). The working lipid and RNA solutions were heated at 37°C for 10 min before being loaded into 3cc luer-lok syringes. 2 mL citrate buffer (pH 6) was loaded in another 3 cc syringe.
  • RNA and the lipids were connected to a T mixer (PEEKTM 500 ⁇ m ID junction, Idex Health Science) using FEP tubing (fluorinated ethylene-propylene; all FEP tubing used had a 2mm internal diameter and a 3mm outer diameter; obtained from Idex Health Science).
  • the outlet from the T mixer was also FEP tubing.
  • the third syringe containing the citrate buffer was connected to a separate piece of tubing. All syringes were then driven at a flow rate of 7 mL/min using a syringe pump. The tube outlets were positioned to collect the mixtures in a 20 mL glass vial (while stirring).
  • the stir bar was taken out and the ethanol/aqueous solution was allowed to equilibrate to room temperature for 1 h. 4 ml of the mixture was loaded into a 5 cc syringe, which was connected to a piece of FEP tubing and in another 5 cc syringe connected to an equal length of FEP tubing, an equal amount of 100 mM citrate buffer (pH 6) was loaded. The two syringes were driven at 7mL/min flow rate using the syringe pump and the final mixture collected in a 20 mL glass vial (while stirring).
  • the mixture collected from the second mixing step were passed through a Mustang Q membrane (an anion-exchange support that binds and removes anionic molecules, obtained from Pall Corporation).
  • a Mustang Q membrane an anion-exchange support that binds and removes anionic molecules, obtained from Pall Corporation.
  • 4 mL of 1 M NaOH, 4 mL of 1 M NaCl and 10 mL of 100 mM citrate buffer (pH 6) were successively passed through it. Liposomes were warmed for 10 min at 37°C before passing through the membrane.
  • liposomes were concentrated to 2 mL and dialyzed against 10-15 volumes of 1X PBS using by tangential flow filtration before recovering the final product.
  • TFF system and hollow fiber filtration membranes were purchased from Spectrum Labs (Rancho Dominguez) and were used according to the manufacturer's guidelines. Polysulfone hollow fiber filtration membranes with a 100 kD pore size cutoff and 8 cm 2 surface area were used. For in vitro and in vivo experiments formulations were diluted to the required RNA concentration with 1X PBS. Further liposome manufacturing methods are disclosed below.
  • Figure 2 shows an example electron micrograph of liposomes prepared by these methods. These liposomes contain encapsulated RNA encoding full-length RSV F antigen. Dynamic light scattering of one batch showed an average diameter of 141nm (by intensity) or 78nm (by number).
  • RNA and RNA concentration were determined by Quant-iT RiboGreen RNA reagent kit (Invitrogen), following manufacturer's instructions. The ribosomal RNA standard provided in the kit was used to generate a standard curve. Liposomes were diluted 10x or 100x in 1X TE buffer (from kit) before addition of the dye. Separately, liposomes were diluted 10x or 100x in 1X TE buffer containing 0.5% Triton X before addition of the dye (to disrupt the liposomes and thus to assay total RNA). Thereafter an equal amount of dye was added to each solution and then ⁇ 180 ⁇ L of each solution after dye addition was loaded in duplicate into a 96 well tissue culture plate. The fluorescence (Ex 485 nm, Em 528 nm) was read on a microplate reader. All liposome formulations were dosed in vivo based on the encapsulated amount of RNA.
  • RNA from liposomes was shown to protect RNA from RNase digestion. Experiments used 3.8mAU of RNase A per microgram of RNA, incubated for 30 minutes at room temperature. RNase was inactivated with Proteinase K at 55°C for 10 minutes. A 1:1 v/v mixture of sample to 25:24:1 v/v/v, phenol:chloroform:isoamyl alcohol was then added to extract the RNA from the lipids into the aqueous phase. Samples were mixed by vortexing for a few seconds and then placed on a centrifuge for 15 minutes at 12k RPM. The aqueous phase (containing the RNA) was removed and used to analyze the RNA.
  • RNA Even after 1 week at 4°C the RNA could be seen without any fragmentation ( Figure 4 , arrow). Protein expression in vivo was unchanged after 6 weeks at 4 °C and one freeze-thaw cycle. Thus liposome-encapsulated RNA is stable.
  • RNA a reporter enzyme SEAP; secreted alkaline phosphatase
  • SEAP secreted alkaline phosphatase
  • Expression levels were measured in sera diluted 1:4 in 1X Phospha-Light dilution buffer using a chemiluminescent alkaline phosphate substrate. 8-10 week old BALB/c mice (5/group) were injected intramuscularly on day 0, 50 ⁇ l per leg with 0.1 ⁇ g or 1 ⁇ g RNA dose. The same vector was also administered without the liposomes (in RNase free 1X PBS) at 1 ⁇ g. Virion-packaged replicons were also tested.
  • Virion-packaged replicons used herein were obtained by the methods of reference 45, where the alphavirus replicon is derived from the mutant VEEV or a chimera derived from the genome of VEEV engineered to contain the 3' UTR of Sindbis virus and a Sindbis virus packaging signal (PS), packaged by co-electroporating them into BHK cells with defective helper RNAs encoding the Sindbis virus capsid and glycoprotein genes.
  • PS Sindbis virus packaging signal
  • encapsulation increased SEAP levels by about 1 ⁇ 2 log at the 1 ⁇ g dose, and at day 6 expression from a 0.1 ⁇ g encapsulated dose matched levels seen with 1 ⁇ g unencapsulated dose.
  • day 3 expression levels exceeded those achieved with VRPs (squares).
  • VRPs squares
  • RNA was formulated in the liposomes relative to the naked RNA control, even at a 10x lower dose. Expression was also higher relative to the VRP control, but the kinetics of expression were very different (see Figure 5 ). Delivery of the RNA with electroporation resulted in increased expression relative to the naked RNA control, but these levels were lower than with liposomes.
  • a replicon was constructed to express full-length F protein from respiratory syncytial virus (RSV). This was delivered naked (1 ⁇ g), encapsulated in liposomes (0.1 or 1 ⁇ g), or packaged in virions (10 6 IU; "VRP") at days 0 and 21.
  • Figure 7 shows anti-F IgG titers 2 weeks after the second dose, and the liposomes clearly enhance immunogenicity.
  • Figure 8 shows titers 2 weeks later, by which point there was no statistical difference between the encapsulated RNA at 0.1 ⁇ g, the encapsulated RNA at 1 ⁇ g, or the VRP group.
  • Neutralisation titers (measured as 60% plaque reduction, "PRNT60") were not significantly different in these three groups 2 weeks after the second dose ( Figure 9 ).
  • Figure 12 shows both IgG and PRNT titers 4 weeks after the second dose.
  • Figure 13 confirms that the RNA elicits a robust CD8 T cell response.
  • liposome-encapsulated RNA induces essentially the same magnitude of immune response as seen with virion delivery.
  • FIG. 11 shows IgG titers in mice receiving the replicon in naked form at 3 different doses, in liposomes at 4 different doses, or as VRP (10 6 IU).
  • the response seen with 1 ⁇ g liposome-encapsulated RNA was statistically insignificant (ANOVA) when compared to VRP, but the higher response seen with 10 ⁇ g liposome-encapsulated RNA was statistically significant (p ⁇ 0.05) when compared to both of these groups.
  • RNA liposome-encapsulated RNA gave much higher anti-F IgG responses (15 days post-second dose) than 0.1 ⁇ g of delivered DNA, and even was more immunogenic than 20 ⁇ g plasmid DNA encoding the F antigen, delivered by electroporation (ElgenTM DNA Delivery System, Inovio).
  • mice showed few visual signs of distress (weight loss, etc.) after receiving liposome-encapsulated RNA replicon, although a transient weight loss of 3-4% was seen after a second dose of 10 ⁇ g RNA. In contrast, delivery of 10 ⁇ g liposome-encapsulated DNA led to 8-10% weight loss.
  • Bone marrow derived dendritic cells were obtained from wild-type mice or the "Resq" ( rsq1 ) mutant strain.
  • the mutant strain has a point mutation at the amino terminus of its TLR7 receptor which abolishes TLR7 signalling without affecting ligand binding [46].
  • the cells were stimulated with replicon RNA formulated with DOTAP, lipofectamine 2000 or inside a liposome.
  • IL-6 and INF ⁇ were induced in WT cells but this response was almost completely abrogated in mutant mice.
  • RNA replicons were shown to induce several serum cytokines within 24 hours of intramuscular injection (IFN- ⁇ , IP-10 (CXCL-10), IL-6, KC, IL-5, IL-13, MCP-1, and MIP-a), whereas only MIP-1 was induced by naked RNA and liposome alone induced only IL-6.
  • IFN- ⁇ was shown to contribute to the immune response to liposome-encapsulated RSV-F-encoding replicon because an anti-IFN ⁇ receptor (IFNAR1) antibody reduced F-specific serum IgG a 10-fold reduction after 2 vaccinations.
  • IFNAR1 anti-IFN ⁇ receptor
  • RNA replicons have generally been seen to elicit a balanced IgG1:IgG2a subtype profile in mice, sometimes with a higher IgG2a/IgG1 ratio than seen with electroporated DNA or with protein/MF59 immunizations (i.e. a Th1-type immune response).
  • lipid expression could be increased 18x by adding 10% 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (DPyPE) to the liposome, 10x by adding 10% 18:2 (cis) phosphatidylcholine, and 900x by instead using RV01.
  • DPyPE 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine
  • the vA317 self-replicating replicon encoding RSV F protein was administered to BALB/c mice, 4 or 8 animals per group, by bilateral intramuscular vaccinations (50 ⁇ L per leg) on days 0 and 21 with the replicon (1 ⁇ g) alone or formulated as liposomes with DlinDMA ("RV01") or DOTAP ("RV13").
  • the RV01 liposomes had 40% DlinDMA, 10% DSPC, 48% cholesterol and 2% PEG-DMG, but with differing amounts of RNA.
  • the RV13 liposomes had 40% DOTAP, 10% DPE, 48% cholesterol and 2% PEG-DMG.
  • naked plasmid DNA (20 ⁇ g) expressing the same RSV-F antigen was delivered either using electroporation or with RV01(10) liposomes (0.1 ⁇ g DNA).
  • RV01(10) liposomes 0.1 ⁇ g DNA.
  • Liposomes were prepared by method (D) or method (B). For some liposomes made by method (D) a double or half amount of RNA was used.
  • the Z average particle diameter, polydispersity index and encapsulation efficiency of the liposomes were as follows: RV Zav (nm) pdI % encapsulation Preparation RV01 (10) 158.6 0.088 90.7 (A) RV01 (08) 156.8 0.144 88.6 (A) RV01 (05) 136.5 0.136 99 (B) RV01 (09) 153.2 0.067 76.7 (A) RV01 (10) 134.7 0.147 87.8 * (A) RV13 (02) 128.3 0.179 97 (A) * For this RV01(10) formulation the nucleic acid was DNA not RNA
  • F-specific serum IgG titers were as follows: RV Day 14 Day 36 Naked DNA plasmid 439 6712 Naked A317 RNA 78 2291 RV01 (10) 3020 26170 RV01 (08) 2326 9720 RV01 (05) 5352 54907 RV01 (09) 4428 51316 RV01 (10) DNA 5 13 RV13 (02) 644 3616
  • the proportion of T cells which are cytokine-positive and specific for RSV F51-66 peptide are as follows, showing only figures which are statistically significantly above zero: RV CD4+CD8- CD4-CD8+ IFN ⁇ IL2 IL5 TNF ⁇ IFN ⁇ IL2 IL5 TNF ⁇ Naked DNA plasmid 0.04 0.07 0.10 0.57 0.29 0.66 Naked A317 RNA 0.04 0.05 0.08 0.57 0.23 0.67 RV01 (10) 0.07 0.10 0.13 1.30 0.59 1.32 RV01 (08) 0.02 0.04 0.06 0.46 0.30 0.51 RV01 (05) 0.08 0.12 0.15 1.90 0.68 1.94 RV01 (09) 0.06 0.08 0.09 1.62 0.67 1.71 RV01 (10) DNA 0.03 0.08 RV13 (02) 0.03 0.04 0.06 1.15 0.41 1.18
  • liposome formulations significantly enhanced immunogenicity relative to the naked RNA controls, as determined by increased F-specific IgG titers and T cell frequencies.
  • RV01 RNA vaccines were more immunogenic than the RV13 vaccine.
  • RV01 has a tertiary amine in the headgroup with a pKa of about 5.8, and also include unsaturated alkyl tails.
  • RV13 has unsaturated alkyl tails but its headgroup has a quaternary amine and is very strongly cationic.
  • the replicon was administered in encapsulated form (with two different purification protocols, 0.1 ⁇ g RNA), or mixed with the liposomes after their formation (a non-encapsulated "lipoplex", 0.1 ⁇ g RNA), or as naked RNA (1 ⁇ g).
  • Figure 10 shows that the lipoplex gave the lowest levels of expression, showing that shows encapsulation is essential for potent expression.
  • RNAs (i) 'vA317' replicon that expresses RSV-F i.e. the surface fusion glycoprotein of RSV; (ii) 'vA17' replicon that expresses GFP; and (iii) 'vA336' that is replication-defective and encodes GFP.
  • RNAs were delivered either naked or with liposomes made by method (D). Empty liposomes were made by method (D) but without any RNA. Four liposome formulations had these characteristics: RNA Particle Size Zav (nm) Polydispersity RNA Encapsulation vA317 155.7 0.113 86.6% vA17 148.4 0.139 92% vA336 145.1 0.143 92.9% Empty 147.9 0.147 -
  • mice 5 animals per group, were given bilateral intramuscular vaccinations (50 ⁇ L per leg) on days 0 and 21 with:
  • Serum was collected for antibody analysis on days 14, 35 and 51.
  • F-specific specific serum IgG titers were measured; if an individual animal had a titer of ⁇ 25 (limit of detection), it was assigned a titer of 5.
  • spleens were harvested from mice at day 51 for T cell analysis, to determine cells which were cytokine-positive and specific for RSV F51-66 peptide (CD4+) or for RSV F peptides F85-93 and F249-258 (CD8+).
  • IgG titers were as follows in the 10 groups and in non-immunised control mice: Day 1 2 3 4 - 14 22 1819 5 5 5 35 290 32533 9 19877 5 51 463 30511 18 20853 5
  • RSV serum neutralization titers at day 51 were as follows: Day 1 2 3 4 51 35 50 24 38
  • Animals showing RSV F-specific CD4+ splenic T cells on day 51 were as follows, where a number (% positive cells) is given only if the stimulated response was statistically significantly above zero: Cytokine 1 2 3 4 IFN- ⁇ 0.04 IL2 0.02 0.06 0.02 IL5 TNF ⁇ 0.03 0.05
  • RNA within the liposomes is necessary for high immunogenicity, as a simple admixture of RNA and the liposomes (group 3) was not immunogenic (in fact, less immunogenic than naked RNA).
  • mice received various combinations of (i) self-replicating RNA replicon encoding full-length RSV F protein (ii) self-replicating GFP-encoding RNA replicon (iii) GFP-encoding RNA replicon with a knockout in nsP4 which eliminates self-replication (iv) full-length RSV F-protein.
  • Results in Figure 16 show that F-specific IgG responses required encapsulation in the liposome rather than mere co-delivery (compare groups C & D).
  • a comparison of groups K, L and M shows that the RNA provided an adjuvant effect against co-delivered protein, and this effect was seen with both replicating and non-replicating RNA.
  • vA142 encodes the full-length wild type surface fusion (F) glycoprotein of RSV but with the fusion peptide deleted, and the 3' end is formed by ribozyme-mediated cleavage. It was tested in three different mouse strains.
  • mice were given bilateral intramuscular vaccinations (50 ⁇ L per leg) on days 0 and 22. Animals were divided into 8 test groups (5 animals per group) and a na ⁇ ve control (2 animals):
  • F-specific serum IgG GMTs were: Day 1 2 3 4 5 6 7 8 14 82 2463 1789 2496 1171 1295 1293 5 35 1538 34181 25605 23579 13718 8887 73809 5
  • F-specific IgG1 and IgG2a titers were as follows: IgG 1 2 3 4 5 6 7 IgG1 94 6238 4836 7425 8288 1817 78604 IgG2a 5386 77064 59084 33749 14437 17624 24
  • RSV serum neutralizing antibody titers at days 35 and 49 were as follows (data are 60% plaque reduction neutralization titers of pools of 2-5 mice, 1 pool per group): Day 1 2 3 4 5 6 7 8 35 ⁇ 20 143 20 101 32 30 111 ⁇ 20 49 ⁇ 20 139 ⁇ 20 83 41 32 1009 ⁇ 20
  • F-specific IgG titers were: Day 1 2 3 4 5 6 7 8 9 14 1140 2133 1026 2792 3045 1330 2975 5 1101 35 1721 5532 3184 3882 9525 2409 39251 5 12139
  • F-specific IgG1 and IgG2a titers were as follows: IgG 1 2 3 4 5 6 7 8 IgG1 66 247 14 328 468 92 56258 79 IgG2a 2170 7685 5055 6161 1573 2944 35 14229
  • RSV serum neutralizing antibody titers at days 35 and 49 were as follows (data are 60% plaque reduction neutralization titers of pools of 2-5 mice, 1 pool per group): Day 1 2 3 4 5 6 7 8 9 35 ⁇ 20 27 29 22 36 ⁇ 20 28 ⁇ 20 ⁇ 20 49 ⁇ 20 44 30 23 36 ⁇ 20 33 ⁇ 20 37
  • F-specific IgG titers were: Day 1 2 3 4 5 6 7 8 9 14 5 2049 1666 1102 298 984 3519 5 806 35 152 27754 19008 17693 3424 6100 62297 5 17249
  • F-specific IgG1 and IgG2a titers were as follows: IgG 1 2 3 4 5 6 7 8 IgG1 5 1323 170 211 136 34 83114 189 IgG2a 302 136941 78424 67385 15667 27085 3800 72727
  • RSV serum neutralizing antibody titers at days 35 and 49 were as follows: Day 1 2 3 4 5 6 7 8 9 35 ⁇ 20 539 260 65 101 95 443 ⁇ 20 595 49 ⁇ 20 456 296 35 82 125 1148 ⁇ 20 387
  • RV01, RV05, RV17; pKa 5.8, 5.85, 6.1 were tested in three different inbred mouse strains.
  • RV01 was more effective than RV17; for BALB/c and C3H strains RV05 was less effective than either RV01 or RV17, but it was more effective in B6 strain.
  • the liposomes were more effective than two cationic nanoemulsions which were tested in parallel.
  • RV01 liposomes with DLinDMA as the cationic lipid were used to deliver RNA replicons encoding cytomegalovirus (CMV) glycoproteins.
  • CMV cytomegalovirus
  • the "vA160" replicon encodes full-length glycoproteins H and L (gH/gL), whereas the "vA322" replicon encodes a soluble form (gHsol/gL).
  • the two proteins are under the control of separate subgenomic promoters in a single replicon; co-administration of two separate vectors, one encoding gH and one encoding gL, did not give good results.
  • mice 10 per group, were given bilateral intramuscular vaccinations (50 ⁇ L per leg) on days 0, 21 and 42 with VRPs expressing gH/gL (1x10 6 IU), VRPs expressing gHsol/gL (1x10 6 IU) and PBS as the controls.
  • Two test groups received 1 ⁇ g of the vA160 or vA322 replicon formulated in liposomes (40% DlinDMA, 10% DSPC, 48% Chol, 2% PEG-DMG; made using method (D) but with 150 ⁇ g RNA batch size).
  • the vA160 liposomes had a Zav diameter of 168nm, a pdI of 0.144, and 87.4% encapsulation.
  • the vA322 liposomes had a Zav diameter of 162nm, a pdI of 0.131, and 90% encapsulation.
  • the replicons were able to express two proteins from a single vector.
  • CMV neutralization titers (the reciprocal of the serum dilution producing a 50% reduction in number of positive virus foci per well, relative to controls) were as follows: gH/gL VRP gHsol/gL VRP gH/gL liposome gHsol/gL liposome 4576 2393 4240 10062
  • RNA expressing either a full-length or a soluble form of the CMV gH/gL complex thus elicited high titers of neutralizing antibodies, as assayed on epithelial cells.
  • the average titers elicited by the liposome-encapsulated RNAs were at least as high as for the corresponding VRPs.
  • RNA replicon was able to express two proteins from a single vector.
  • the RNA replicon gave a 3wp3 titer of 11457, compared to 5516 with VRPs.
  • the vA526 replicon expresses the CMV pentameric complex (gH-gL-UL128-UL130-UL-131) under the control of three subgenomic promoters: the first drives the expression of gH; the second drives expression of gL; the third drives the expression of the UL128-2A-UL130-2A-UL131 polyprotein, which contains two 2A cleavage sites between the three UL genes.
  • the vA527 replicon expresses the CMV pentameric complex via three subgenomic promoters and two IRESs: the first subgenomic promoter drives the expression of gH; the second subgenomic promoter drives expression of gL; the third subgenomic promoter drives the expression of the UL128; UL130 is under the control of an EMCV IRES; UL131 is under control of an EV71 IRES.
  • H liposome
  • VRPs VRPs.
  • mice 10 groups of 10 animals, were given bilateral intramuscular vaccinations (50 ⁇ L per leg) on days 0, 21 and 42 with:
  • Sera were collected for immunological analysis on days 21 (3wp1), 42 (3wp2) and 63 (3wp3).
  • CMV serum neutralization titers on days 21, 42 and 63 were: Vaccine Group 3wP1 3wp2 3wp3 1 126 6296 26525 2 N/A N/A 6769 3 N/A 3442 7348 4 N/A N/A 2265 5 347 9848 42319 6 179 12210 80000 7 1510 51200 130000 8 N/A N/A 845 9 N/A N/A 228 10 N/A N/A 413
  • self-replicating RNA can be used to express multiple antigens from a single vector and to raise a potent and specific immune response.
  • the replicon can express five antigens (CMV pentamric complex (gH-gL-UL128-UL130-UL-131) and raise a potent immune response.
  • CMV pentamric complex gH-gL-UL128-UL130-UL-131
  • Self-replicating RNA delivered in liposomes was able to elicit high titers of neutralizing antibody, as assayed on epithelial cells, at all time points assayed (3wp1, 3wp2, and 3wp3). These responses were superior to the corresponding VRPs and to cationic nanoemulsions.
  • Hydrodynamic delivery employs the force generated by the rapid injection of a large volume of solution to overcome the physical barriers of cell membranes which prevent large and membrane-impermeable compounds from entering cells. This phenomenon has previously been shown to be useful for the intracellular delivery of DNA vaccines.
  • a typical mouse delivery volume for intramuscular injection is 50 ⁇ l into the hind leg, which is a relatively high volume for a mouse leg muscle.
  • a human intramuscular dose of ⁇ 0.5ml is relatively small. If immunogenicity in mice would be volume-dependent then the replicon vaccines' efficacy might be due, at least in part, on hydrodynamic forces, which would not be encouraging for use of the same vaccines in humans and larger animals.
  • the vA317 replicon was delivered to BALB/c mice, 10 per group, by bilateral intramuscular vaccinations (5 or 50 per leg) on day 0 and 21:
  • F-specific serum IgG GMTs were: Day 1 2 3 4 14 42 21 2669 2610 35 241 154 17655 18516
  • vA311 A self-replicating RNA replicon (“vA311”) that expresses a luciferase reporter gene (luc) was used for studying the kinetics of protein expression after injection.
  • mice Prior to vaccination mice were depilated. Mice were anesthetized (2% isoflurane in oxygen), hair was first removed with an electric razor and then chemical Nair. Bioluminescence data was then acquired using a Xenogen IVIS 200 imaging system (Caliper Life Sciences) on days 3, 7, 14, 21, 28, 35, 42, 49, 63 and 70. Five minutes prior to imaging mice were injected intraperitoneally with 8 mg/kg of luciferin solution. Animals were then anesthetized and transferred to the imaging system. Image acquisition times were kept constant as bioluminescence signal was measured with a cooled CCD camera.
  • luciferase-expressing cells were seen to remain primarily at the site of RNA injection, and animals imaged after removal of quads showed no signal.
  • luciferase expression was measured as average radiance over a period of 70 days (p/s/cm 2 /sr), and results were as follows for the 5 groups: Days 1 2 3 4 5 3 8.69E+07 3.33E+06 2.11E+06 9.71E+06 1.46E+07 7 1.04E+08 8.14E+06 1.83E+07 5.94E+07 1.64E+07 14 8.16E+07 2.91E+06 9.22E+06 3.48E+07 8.49E+05 21 1.27E+07 3.13E+05 6.79E+04 5.07E+05 6.79E+05 28 1.42E+07 6.37E+05 2.36E+04 4.06E+03 2.00E+03 35 1.21E+07 6.12E+05 2.08E+03 42 1.49E+07 8.70E+05 49 1.17E+07 2.04E+05 63 9.69E+06 1.72E+03 70 9.29E+06
  • RNA formulated with cationic nanoemulsions showed measurable bioluminescence at day 3, which peaked at day 7 and then reduced to background levels by days 28 to 35.
  • RNA When formulated in liposomes the RNA showed measurable bioluminescence at day 3, which peaked at day 7 and reduced to background levels by day 63.
  • RNA delivered using VRPs showed enhanced bioluminescence at day 21 when compared to the formulated RNA, but expression had reduced to background levels by day 28.
  • Electroporated DNA showed the highest level of bioluminescence at all time points measured and levels of bioluminescence did not reduce to background levels within the 70 days of the experiment.
  • Liposome-encapsulated RNA encoding HIV gp140 was delivered to mice intramuscularly, intradermally, or subcutaneously. All three routes led to high serum IgG levels of HIV-specific antibodies ( Figure 15 ), exceeding titers seen in response to electroporated intramuscular DNA.
  • vA317 expresses full-length RSV-F
  • vA318 expresses truncated (transmembrane and cytoplasmic tail removed) RSV-F
  • vA142 expresses RSV-F with its fusion peptide deleted
  • vA140 expresses the truncated RSV-F also without its peptide.
  • Cotton rats, 4 to 8 animals per group, were given intramuscular vaccinations (100 ⁇ L in one leg) on days 0 and 21 with the four different replicons at two doses (1.0 and 0.1 ⁇ g) formulated in liposomes made by method (D), but with a 150 ⁇ g RNA batch size.
  • Control groups received a RSV-F subunit protein vaccine (5 ⁇ g) adjuvanted with alum (8 animals/group), VRPs expressing full-length RSV-F (1x10 6 IU, 8 animals/group), or na ⁇ ve control (4 animals/group). Serum was collected for antibody analysis on days 0, 21 and 34.
  • F-specific serum IgG titers and RSV serum neutralizing antibody titers on day 21 and 34 were: Group IgG, day 21 IgG, day 34 NT, day 21 NT, day 34 1 ⁇ g vA317 915 2249 115 459 0.1 ⁇ g vA317 343 734 87 95 1 ⁇ g vA318 335 1861 50 277 0.1 ⁇ g vA318 129 926 66 239 1 ⁇ g vA142 778 4819 92 211 0.1 ⁇ g vA142 554 2549 78 141 1 ⁇ g vA140 182 919 96 194 0.1 ⁇ g vA140 61 332 29 72 5 ⁇ g F trimer subunit/alum 13765 86506 930 4744 1x10 6 IU VRP-F full 1877 19179 104 4528 Naive 5 5 10 15
  • All four replicons evaluated in this study were immunogenic in cotton rats when delivered by liposome, although serum neutralization titers were at least ten-fold lower than those induced by adjuvanted protein vaccines or by VRPs.
  • the liposome/RNA vaccines elicited serum F-specific IgG and RSV neutralizing antibodies after the first vaccination, and a second vaccination boosted the response effectively.
  • F-specific IgG titers after the second vaccination with 1 ⁇ g replicon were 2- to 3-fold higher than after the second vaccination with 0.1 ⁇ g replicon.
  • the four replicons elicited comparable antibody titers, suggesting that full length and truncated RSV-F, each with or without the fusion peptide, are similarly immunogenic in cotton rats.
  • Cotton rats 2-8 animals per group, were given intramuscular vaccinations (100 ⁇ L in one leg) on days 0 and 21 with the replicons (0.1 or 1 ⁇ g) encapsulated in RV01 liposomes made by method (D) but with a 150 ⁇ g RNA batch size.
  • Control groups received the RSV-F subunit protein vaccine (5 ⁇ g) adjuvanted with alum or VRPs expressing full-length RSV-F (1x10 6 IU, 8 animals/group). All these animals received a third vaccination (day 56) with RSV-F subunit protein vaccine (5 ⁇ g) adjuvanted with alum.
  • F-specific serum IgG titers were as follows: Day 21 Day 35 Day 56 Day 70 1 ⁇ g vA318 260 1027 332 14263 0.1 ⁇ g vA318 95 274 144 2017 1 ⁇ g vA142 483 1847 1124 11168 0.1 ⁇ g vA142 314 871 418 11023 1 ⁇ g vA317 841 4032 1452 13852 1x10 6 VRP (F-full) 2075 3938 1596 14574 5 ⁇ g F trimer subunit/alum 12685 54526 25846 48864 Naive 5 5 5 5 5
  • Serum neutralisation titers were as follows (60% RSV neutralization titers for 2 pools of 3-4 animals per group, GMT of these 2 pools per group): Day 21 Day 35 Day 56 Day 70 1 ⁇ g vA318 58 134 111 6344 0.1 ⁇ g vA318 41 102 63 6647 1 ⁇ g vA142 77 340 202 5427 0.1 ⁇ g vA142 35 65 56 2223 1 ⁇ g vA317 19 290 200 4189 1x10 6 VRP (F-full) 104 1539 558 2876 5 ⁇ g F trimer subunit/alum 448 4457 1630 3631 Naive 10 10 10 10
  • Serum titers and neutralising titers for the extra group were as follows: Day 14 21 28 35 42 56 70 IgG 397 561 535 501 405 295 3589 NT 52 82 90 106 80 101 1348
  • the replicons are confirmed as immunogenic in cotton rats, eliciting serum F-specific IgG and RSV neutralizing antibodies after the first vaccination.
  • a second vaccination boosted the responses effectively.
  • F-specific IgG titers after the second vaccination with 1.0 ⁇ g replicon were 1.5 to 4-fold higher than after the second vaccination with 0.1 ⁇ g replicon.
  • the third vaccination did not boost titers in cotton rats previously vaccinated with F trimer subunit + alum, but it did provide a large boost to titers in cotton rats previously vaccinated with replicon.
  • the RSV serum neutralization titers after two replicon vaccinations followed by protein boost were equal to or greater than titers induced by two or three sequential protein vaccinations.
  • F-specific serum IgG and RSV neutralization titers induced by a single vaccination reached their peak around day 21 and were maintained through at least day 56 (50-70% drop in F-specific IgG titer, little change in RSV neutralization titer).
  • a homologous second vaccination was given to these animals on day 56, and boosted antibody titers to a level at least equal to that achieved when the second vaccination was administered on day 21.
  • vA368 replicon encodes the full-length wild type surface fusion glycoprotein of RSV with the fusion peptide deleted, with expression driven by the EV71 IRES.
  • a control group received 5 ⁇ g alum-adjuvanted protein, and a na ⁇ ve control group was also included.
  • RNA vaccine reduced the lung viral load by over three logs, from approximately 10 6 PFU/g in unvaccinated control cotton rats to less than 10 3 PFU/g in vaccinated cotton rats.
  • RNA vaccines encoded human RSV F whereas the "Triangle 4" vaccine contains bovine RSV F, but the RSV F protein is highly conserved between BRSV and HRSV.
  • the liposomes were made by method (E), except a 1.5 mg RNA batch size was used.
  • Serum was collected for antibody analysis on days 0, 14, 21, 35, 42, 56, 63, 86, 100, 107, 114, 121, 128, 135, 146, 160, 167, 174, 181, 188, 195, and 202. If an individual animal had a titer below the limit of detection it was assigned a titer of 5
  • Figure 14A shows F-specific IgG titers over the first 63 days.
  • the RNA replicon was immunogenic in the cows via liposomes, although it gave lower titers than the licensed vaccine. All vaccinated cows showed F-specific antibodies after the second dose, and titers were very stable from the period of 2 to 6 weeks after the second dose (and were particularly stable for the RNA vaccines).
  • Figure 14B shows F-specific serum IgG titers (GMT) over 210 days, and measured values up to day 202 were as follows: D0 3wp1 D21 2wp2 D35 5wp2 D56 ⁇ 9wp2 D86 2wp3 D100 5wp3 D121 8wp3 D146 2wp4 D160 5wp4 D181 8wp4 D202 PBS 5 5 5 5 5 5 5 5 46 98 150 Liposome 5 5 12 11 20 768 428 74 20774 7022 2353 Triangle 4 5 5 5 1784 721 514 3406 2786 336 13376 4775 2133
  • RSV serum neutralizing antibody titers were as follows: D0 2wp2 D35 5wp2 D56 2wp3 D100 3wp3 D107 4wp3 D114 8wp3 D146 2wp4 D160 3wp4 D167 4wp4 D174 PBS 12 10 10 14 18 20 14 10 10 10 Liposome 13 10 10 20 13 17 13 47 26 21 Triangle 4 12 15 13 39 38 41 13 24 26 15
  • the material used for the second liposome dose was not freshly prepared, and the same lot of RNA showed a decrease in potency in a mouse immunogenicity study. Therefore it is possible that the vaccine would have been more immunogenic if fresh material had been used for all vaccinations.
  • MF59-adjuvanted RSV-F was able to boost the IgG response in all previously vaccinated calves, and to boost complement-independent neutralization titers of calves previously vaccinated with RNA.
  • RNA vaccines in large animals is particularly important in light of the loss in potency observed previously with DNA-based vaccines when moving from small animal models to larger animals and humans.
  • a typical dose for a cow DNA vaccine would be 0.5-1 mg [47,48] and so it is very encouraging that immune responses were induced with only 66 ⁇ g of RNA.
  • Table 1 useful phospholipids DDPC 1,2-Didecanoyl-sn-Glycero-3-phosphatidylcholine DEPA 1,2-Dierucoyl-sn-Glycero-3-Phosphate DEPC 1,2-Erucoyl-sn-Glycero-3-phosphatidylcholine DEPE 1,2-Dierucoyl-sn-Glycero-3-phosphatidylethanolamine DEPG 1,2-Dierucoyl-sn-Glycero-3[Phosphatidyl-rac-(1-glycerol%) DLOPC 1,2-Linoleoyl-sn-Glycero-3-phosphatidylcholine DLPA 1,2-Dilauroyl-sn-Glycero-3-Phosphate DLPC 1,2-Dilauroyl-sn-Glycero-3-phosphatidylcholine DLPE 1,2-Dilauroyl-sn-Glycero-3-phosphatidy

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Virology (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Mycology (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Plant Pathology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Dispersion Chemistry (AREA)
  • Pulmonology (AREA)
  • Dermatology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Manufacturing Of Micro-Capsules (AREA)

Claims (13)

  1. Non-Virion-Partikel, das kein Proteinkapsid umfasst, für die In-vivo-Verabreichung von RNA an eine Wirbeltierzelle, wobei (a) das Partikel ein Liposom ist und ein Abgabematerial umfasst, das ein selbstreplizierendes RNA-Molekül verkapselt, das ein Immunogen kodiert, wobei das Immunogen ein Oberflächenpolypeptid ist und eine Immunreaktion in vivo gegen ein Bakterium, einen Virus, einen Pilz oder einen Parasit auslösen kann und (b) die RNA keine modifizierten Nukleotide umfasst und gegebenenfalls eine 5'-Kappe umfasst.
  2. Partikel nach Anspruch 1, wobei das Liposom ein Lipid mit einer kationischen Kopfgruppe umfasst.
  3. Partikel nach Anspruch 1, wobei das Liposom ein Lipid mit einer zwitterionischen Kopfgruppe umfasst.
  4. Partikel nach einem der Ansprüche 1 bis 3, wobei das Liposom einen Durchmesser im Bereich von 50-220 nm aufweist.
  5. Partikel nach irgendeinem vorhergehenden Anspruch, wobei das selbst-replizierende RNA-Molekül (i) eine RNA-abhängige RNA-Polymerase, die RNA von dem selbstreplizierenden RNA-Molekül transkribieren kann und (ii) ein Immunogen kodiert.
  6. Partikel nach Anspruch 5, wobei die Polymerase eine Alphavirus-Replikase ist.
  7. Partikel nach Anspruch 5 oder Anspruch 6, wobei das RNA-Molekül zwei offene Leseraster aufweist, wobei das erste davon eine Alphavirus-Replikase kodiert und das zweite davon das Immunogen kodiert.
  8. Partikel nach Anspruch 7, wobei das RNA-Molekül zusätzliche offene Leseraster, beispielsweise zum Kodieren von weiteren immunogenen oder zusätzlichen Polypeptiden aufweist.
  9. Partikel nach irgendeinem vorhergehenden Anspruch, wobei das RNA-Molekül 5000-25000 Nukleotide lang ist.
  10. Partikel nach irgendeinem vorhergehenden Anspruch, wobei das Immunogen eine Immunreaktion in vivo gegen:
    (a) respiratorisches Syncytialvirus-Glykoprotein F;
    (b) einen Virus, der Fische infiziert, wie Virus der infektiösen Lachsanämie (ISAV), Lachs-Bauchspeicheldrüsen-Krankheitsvirus (SPDV), infektiöser Pankreasnekrosevirus (IPNV), Channel-Catfish-Virus (CCV), Fisch-Lyphocystisdisease-Virus (FLDV), infektiöser hämatopoetischer Nekrosevirus (IHNV), Koi-Herpesvirus, Lachs-Picorna-artiger Virus (auch als Picorna-artiger Virus von atlantischem Lachs bekannt), Binnenlachsvirus (LSV), atlantischer Lachs-Rotavirus (ASR), Forelle-Strawberrydisease-Virus (TSD), Silberlachstumorvirus (CSTV) und viraler hämorrhagischer Septikämie-Virus (VHSV); (c) Orthomyxovirus, wie Influenza A-, -B- und -C-Virus; oder (d) Herpesvirus, wie Herpes simplex-Viren (HSV), Varicella-Zoster-Virus (VZV), Epstein-Barr-Virus (EBV), Cytomegalovirus (CMV), humaner Herpesvirus 6 (HHV6), humaner Herpesvirus 7 (HHV7) und humaner Herpesvirus 8 (HHV8) auslösen kann.
  11. Pharmazeutische Zusammensetzung, umfassend ein Partikel nach irgendeinem vorhergehenden Anspruch.
  12. Abgabevorrichtung, enthaltend die pharmazeutische Zusammensetzung nach Anspruch 11.
  13. Partikel nach irgendeinem der Ansprüche 1-10 oder pharmazeutische Zusammensetzung nach Anspruch 11 oder Abgabevorrichtung nach Anspruch 12 für die Verwendung in einem Verfahren zur Erzeugung einer Schutzimmunantwort in einem Wirbeltier, umfassend den Schritt der Verabreichung einer wirksamen Menge des genannten Partikels oder der genannten pharmazeutischen Zusammensetzung an das Wirbeltier.
EP11741348.4A 2010-07-06 2011-07-06 Virion-artige transferpartikel für selbstreplizierende rna-moleküle Revoked EP2590676B1 (de)

Priority Applications (6)

Application Number Priority Date Filing Date Title
RS20160879A RS55260B1 (sr) 2010-07-06 2011-07-06 Čestice slične virionu za dopremanje samoreplicirajućih rnk molekula
EP22200367.5A EP4180057A1 (de) 2010-07-06 2011-07-06 Virionabgabepartikel für selbstreplizierende rna-moleküle
EP16184271.1A EP3115061A1 (de) 2010-07-06 2011-07-06 Virionabgabepartikel für selbstreplizierende rna-moleküle
EP21204155.2A EP4005592B1 (de) 2010-07-06 2011-07-06 Virionabgabepartikel für selbstreplizierende rna-moleküle
SI201130994A SI2590676T1 (sl) 2010-07-06 2011-07-06 Virionu podobni delci za dajanje za samorepliciranje RNA molekul
HRP20161352TT HRP20161352T1 (hr) 2010-07-06 2016-10-17 Čestice nalik na virione za unos samoreplicirajućih molekula rna

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US36182810P 2010-07-06 2010-07-06
PCT/US2011/043103 WO2012006376A2 (en) 2010-07-06 2011-07-06 Virion-like delivery particles for self-replicating rna molecules

Related Child Applications (3)

Application Number Title Priority Date Filing Date
EP21204155.2A Division EP4005592B1 (de) 2010-07-06 2011-07-06 Virionabgabepartikel für selbstreplizierende rna-moleküle
EP22200367.5A Division EP4180057A1 (de) 2010-07-06 2011-07-06 Virionabgabepartikel für selbstreplizierende rna-moleküle
EP16184271.1A Division EP3115061A1 (de) 2010-07-06 2011-07-06 Virionabgabepartikel für selbstreplizierende rna-moleküle

Publications (2)

Publication Number Publication Date
EP2590676A2 EP2590676A2 (de) 2013-05-15
EP2590676B1 true EP2590676B1 (de) 2016-08-17

Family

ID=44629863

Family Applications (4)

Application Number Title Priority Date Filing Date
EP11741348.4A Revoked EP2590676B1 (de) 2010-07-06 2011-07-06 Virion-artige transferpartikel für selbstreplizierende rna-moleküle
EP22200367.5A Pending EP4180057A1 (de) 2010-07-06 2011-07-06 Virionabgabepartikel für selbstreplizierende rna-moleküle
EP16184271.1A Withdrawn EP3115061A1 (de) 2010-07-06 2011-07-06 Virionabgabepartikel für selbstreplizierende rna-moleküle
EP21204155.2A Active EP4005592B1 (de) 2010-07-06 2011-07-06 Virionabgabepartikel für selbstreplizierende rna-moleküle

Family Applications After (3)

Application Number Title Priority Date Filing Date
EP22200367.5A Pending EP4180057A1 (de) 2010-07-06 2011-07-06 Virionabgabepartikel für selbstreplizierende rna-moleküle
EP16184271.1A Withdrawn EP3115061A1 (de) 2010-07-06 2011-07-06 Virionabgabepartikel für selbstreplizierende rna-moleküle
EP21204155.2A Active EP4005592B1 (de) 2010-07-06 2011-07-06 Virionabgabepartikel für selbstreplizierende rna-moleküle

Country Status (22)

Country Link
US (2) US11291635B2 (de)
EP (4) EP2590676B1 (de)
JP (1) JP6061849B2 (de)
CN (2) CN103052400B (de)
AU (1) AU2011276232B2 (de)
BR (1) BR112013000392B8 (de)
CA (1) CA2804494A1 (de)
CY (1) CY1118080T1 (de)
DK (1) DK2590676T3 (de)
ES (2) ES2934240T3 (de)
FI (1) FI4005592T3 (de)
HR (2) HRP20221522T1 (de)
HU (2) HUE060788T2 (de)
LT (2) LT4005592T (de)
MX (1) MX342608B (de)
PL (2) PL4005592T3 (de)
PT (2) PT2590676T (de)
RS (2) RS63817B1 (de)
RU (1) RU2013104890A (de)
SI (2) SI2590676T1 (de)
SM (1) SMT201600386B (de)
WO (1) WO2012006376A2 (de)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2729126B1 (de) 2011-07-06 2020-12-23 GlaxoSmithKline Biologicals SA Liposomen mit nützlichem n:p-verhältnis zur freisetzung von rna-molekülen
US11291635B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biological Sa Virion-like delivery particles for self-replicating RNA molecules
US11291682B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US20220125723A1 (en) 2010-07-06 2022-04-28 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US11504421B2 (en) 2017-05-08 2022-11-22 Gritstone Bio, Inc. Alphavirus neoantigen vectors
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11771747B2 (en) 2020-08-06 2023-10-03 Gritstone Bio, Inc. Multiepitope vaccine cassettes
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof

Families Citing this family (163)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6025721B2 (ja) 2010-07-06 2016-11-16 ノバルティス アーゲー カチオン性水中油型エマルジョン
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
EP4119155A1 (de) 2010-08-31 2023-01-18 GlaxoSmithKline Biologicals S.A. Pegylierte liposomen zur auslieferung von immunogen-kodierende rns
PL2611467T3 (pl) * 2010-08-31 2022-08-16 Glaxosmithkline Biologicals Sa Małe liposomy do dostarczania rna kodującego immunogen
HUE058896T2 (hu) 2010-10-01 2022-09-28 Modernatx Inc N1-metil-pszeudo-uracilt tartalmazó ribonukleinsavak és azok felhasználásai
TR201903651T4 (tr) * 2010-10-11 2019-04-22 Glaxosmithkline Biologicals Sa Antijen uygulama platformları.
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
JP6184945B2 (ja) 2011-06-08 2017-08-23 シャイアー ヒューマン ジェネティック セラピーズ インコーポレイテッド mRNA送達のための脂質ナノ粒子組成物および方法
RU2606846C2 (ru) 2011-07-06 2017-01-10 Новартис Аг Эмульсии типа "масло в воде", которые содержат нуклеиновые кислоты
ES2702318T3 (es) 2011-07-06 2019-02-28 Glaxosmithkline Biologicals Sa Emulsiones catiónicas de aceite en agua
MX366055B (es) * 2011-08-31 2019-06-26 Novartis Ag Liposomas pegilados para admistracion de acido ribonucleico (arn) que codifica para inmunogeno.
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
RS62993B1 (sr) 2011-10-03 2022-03-31 Modernatx Inc Modifikovani nukleozidi, nukleotidi, i nukleinske kiseline, i njihove upotrebe
AU2012322704B2 (en) * 2011-10-11 2017-09-07 Novartis Ag Recombinant self-replicating polycistronic RNA molecules
CN104114572A (zh) 2011-12-16 2014-10-22 现代治疗公司 经修饰的核苷、核苷酸和核酸组合物
DE18200782T1 (de) 2012-04-02 2021-10-21 Modernatx, Inc. Modifizierte polynukleotide zur herstellung von proteinen im zusammenhang mit erkrankungen beim menschen
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
JP2015518705A (ja) 2012-04-02 2015-07-06 モデルナ セラピューティクス インコーポレイテッドModerna Therapeutics,Inc. ヒト疾患に関連する生物製剤およびタンパク質の産生のための修飾ポリヌクレオチド
EA201492055A1 (ru) 2012-06-08 2015-11-30 Шир Хьюман Дженетик Терапис, Инк. ИНГАЛЯЦИОННАЯ ДОСТАВКА мРНК В НЕЛЕГОЧНЫЕ КЛЕТКИ-МИШЕНИ
WO2014028429A2 (en) 2012-08-14 2014-02-20 Moderna Therapeutics, Inc. Enzymes and polymerases for the synthesis of rna
EP4074834A1 (de) 2012-11-26 2022-10-19 ModernaTX, Inc. Am kettenende modifizierte rna
WO2014108515A1 (en) * 2013-01-10 2014-07-17 Novartis Ag Influenza virus immunogenic compositions and uses thereof
JP2016504050A (ja) 2013-01-17 2016-02-12 モデルナ セラピューティクス インコーポレイテッドModerna Therapeutics,Inc. 細胞表現型の改変のためのシグナルセンサーポリヌクレオチド
EP2968391A1 (de) 2013-03-13 2016-01-20 Moderna Therapeutics, Inc. Langlebige polynukleotidmoleküle
EP2971010B1 (de) 2013-03-14 2020-06-10 ModernaTX, Inc. Formulierung und abgabe von modifizierten nukleosid-, nukleotid- und nukleinsäurezusammensetzungen
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
CA2906281A1 (en) 2013-03-15 2014-09-18 Glaxosmithkline Biologicals Sa Rna purification methods
EP3971287A1 (de) 2013-07-11 2022-03-23 ModernaTX, Inc. Zusammensetzungen mit synthetischen polynukleotiden zur codierung von crispr-verwandten proteinen und synthetischen sgrnas und verfahren zur verwendung
AU2014315287A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
EP3041938A1 (de) 2013-09-03 2016-07-13 Moderna Therapeutics, Inc. Kreisförmige polynukleotide
EP3052106A4 (de) 2013-09-30 2017-07-19 ModernaTX, Inc. Polynukleotide zur codierung immunmodulierender polypeptide
MX2016004249A (es) 2013-10-03 2016-11-08 Moderna Therapeutics Inc Polinulcleotidos que codifican el receptor de lipoproteina de baja densidad.
WO2015126477A1 (en) * 2013-11-08 2015-08-27 Tyrx, Inc. Polymeric drug delivery system for treating surgical complications
EP3798229A1 (de) * 2014-03-25 2021-03-31 Yale University Verwendung von migrationshemmungsfaktoren parasitärer makrophagen
PT3134131T (pt) 2014-04-23 2022-03-24 Modernatx Inc Vacinas de ácidos nucleicos
EP2974739A1 (de) 2014-07-15 2016-01-20 Novartis AG RSVF-Trimerisierungsdomänen
WO2015189425A1 (en) 2014-06-13 2015-12-17 Glaxosmithkline Biologicals Sa Immunogenic combinations
WO2016011226A1 (en) 2014-07-16 2016-01-21 Moderna Therapeutics, Inc. Chimeric polynucleotides
US20170210788A1 (en) 2014-07-23 2017-07-27 Modernatx, Inc. Modified polynucleotides for the production of intrabodies
EP3061826A1 (de) 2015-02-27 2016-08-31 Novartis AG Flavivirus-replikons
US10293058B2 (en) 2015-04-22 2019-05-21 Curevac Ag RNA containing composition for treatment of tumor diseases
TW201718638A (zh) 2015-07-21 2017-06-01 現代治療公司 傳染病疫苗
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
SI3350157T1 (sl) 2015-09-17 2022-04-29 Modernatx, Inc. Sestave za doziranje terapevtskih sredstev v celice
EP3359670B2 (de) 2015-10-05 2024-02-14 ModernaTX, Inc. Verfahren zur therapeutischen verabreichung messenger-ribonukleinsäure-arzneimitteln
KR20180094859A (ko) 2015-10-22 2018-08-24 모더나티엑스, 인크. 수두 대상포진 바이러스 (vzv)를 위한 핵산 백신
US20180289792A1 (en) 2015-10-22 2018-10-11 ModernaTX. Inc. Sexually transmitted disease vaccines
CA3002922A1 (en) * 2015-10-22 2017-04-27 Modernatx, Inc. Human cytomegalovirus vaccine
MA46023A (fr) * 2015-10-22 2019-07-03 Modernatx Inc Vaccin contre le virus de la grippe à large spectre
EP3364950A4 (de) 2015-10-22 2019-10-23 ModernaTX, Inc. Impfstoffe gegen tropenkrankheiten
EP3365009A4 (de) * 2015-10-22 2019-07-03 ModernaTX, Inc. Impfstoffe gegen herpes-simplex-virus
BR112018008102A2 (pt) * 2015-10-22 2018-11-06 Modernatx Inc vacina de vírus sincicial respiratório
EP3964200A1 (de) 2015-12-10 2022-03-09 ModernaTX, Inc. Zusammensetzungen und verfahren zur abgabe von therapeutischen wirkstoffen
JP7114465B2 (ja) 2015-12-22 2022-08-08 モデルナティエックス インコーポレイテッド 薬剤の細胞内送達のための化合物および組成物
PT3394093T (pt) 2015-12-23 2022-05-30 Modernatx Inc Métodos de utilização de polinucleotídeos que codificam ligandos ox40
EP3400023A1 (de) 2016-01-10 2018-11-14 ModernaTX, Inc. Therapeutische mrnas zur codierung von anti-ctla-4-antikörpern
BR112018014109A2 (pt) 2016-01-11 2018-12-11 Verndari Inc composições de microagulhas e métodos de uso das mesmas
WO2017162265A1 (en) 2016-03-21 2017-09-28 Biontech Rna Pharmaceuticals Gmbh Trans-replicating rna
BE1024796B9 (fr) 2016-06-02 2019-01-08 Glaxosmithkline Biologicals Sa Constructions antigeniques du virus zika
WO2018060288A1 (en) 2016-09-29 2018-04-05 Glaxosmithkline Biologicals S.A. Compositions and methods of treatment of persistent hpv infection
GB201616904D0 (en) 2016-10-05 2016-11-16 Glaxosmithkline Biologicals Sa Vaccine
MA46584A (fr) 2016-10-21 2019-08-28 Modernatx Inc Vaccin contre le cytomégalovirus humain
WO2018089540A1 (en) 2016-11-08 2018-05-17 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
WO2018089851A2 (en) 2016-11-11 2018-05-17 Modernatx, Inc. Influenza vaccine
EP4043031A3 (de) 2016-11-17 2022-11-23 GlaxoSmithKline Biologicals SA Antigenkonstrukte gegen zika-virus
MA50335A (fr) 2016-12-08 2020-08-19 Modernatx Inc Vaccins à acide nucléique contre des virus respiratoires
KR20190124750A (ko) 2017-02-28 2019-11-05 사노피 치료적 rna
MA47787A (fr) 2017-03-15 2020-01-22 Modernatx Inc Vaccin contre le virus respiratoire syncytial
US11969506B2 (en) 2017-03-15 2024-04-30 Modernatx, Inc. Lipid nanoparticle formulation
MA52262A (fr) 2017-03-15 2020-02-19 Modernatx Inc Vaccin à large spectre contre le virus de la grippe
US11752206B2 (en) 2017-03-15 2023-09-12 Modernatx, Inc. Herpes simplex virus vaccine
WO2018170270A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Varicella zoster virus (vzv) vaccine
DK3596041T3 (da) 2017-03-15 2023-01-23 Modernatx Inc Forbindelse og sammensætninger til intracellulær afgivelse af terapeutiske midler
ES2911186T3 (es) 2017-03-15 2022-05-18 Modernatx Inc Formas cristalinas de aminolípidos
ES2952779T3 (es) 2017-05-18 2023-11-06 Modernatx Inc ARN mensajero modificado que comprende elementos de ARN funcionales
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
EP3638292A1 (de) 2017-06-14 2020-04-22 ModernaTX, Inc. Für koagulationsfaktor viii kodierende polynukleotide
EP3638776A1 (de) 2017-06-14 2020-04-22 Technische Universität Dresden Verfahren und mittel zur genetischen veränderung von genomen unter verwendung von designer-dna-rekombinatiosenzymen
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
US11235051B2 (en) 2017-07-28 2022-02-01 Janssen Vaccines & Prevention B.V. Methods and compositions for heterologous repRNA immunizations
US11639329B2 (en) 2017-08-16 2023-05-02 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
US10653767B2 (en) 2017-09-14 2020-05-19 Modernatx, Inc. Zika virus MRNA vaccines
EP3461497A1 (de) 2017-09-27 2019-04-03 GlaxoSmithKline Biologicals S.A. Virale antigene
EP3714048A1 (de) 2017-11-22 2020-09-30 Modernatx, Inc. Für ornithintranscarbamylase codierende polynukleotide zur behandlung von harnstoffzyklusstörungen
EP3714045A1 (de) 2017-11-22 2020-09-30 Modernatx, Inc. Für propionyl-coa-carboxylase-alpha- und beta-untereinheiten codierende polynukleotide zur behandlung von propionazidämie
EP3714047A2 (de) 2017-11-22 2020-09-30 ModernaTX, Inc. Polynukleotide codierend für phenylalaninhydroxylase zur behandlung von phenylketonurie
US11020476B2 (en) 2017-12-19 2021-06-01 Janssen Sciences Ireland Unlimited Company Methods and compositions for inducing an immune response against Hepatitis B Virus (HBV)
US11389531B2 (en) 2017-12-19 2022-07-19 Janssen Sciences Ireland Unlimited Company Methods and apparatus for the delivery of hepatitis B virus (HBV) vaccines
EA202091513A1 (ru) 2017-12-19 2020-09-09 Янссен Сайенсиз Айрлэнд Анлимитед Компани Вакцины против вируса гепатита b (hbv) и их применение
BR112020012361A2 (pt) 2017-12-20 2020-11-24 Glaxosmithkline Biologicals S.A. constructos de antígeno do vírus epstein-barr
CN111818943A (zh) 2018-01-04 2020-10-23 伊科尼克治疗公司 抗组织因子抗体、抗体-药物缀合物及相关方法
EP3735270A1 (de) 2018-01-05 2020-11-11 Modernatx, Inc. Für anti-chikungunya-virus-antikörper codierende polynukleotide
MA54676A (fr) 2018-01-29 2021-11-17 Modernatx Inc Vaccins à base d'arn contre le vrs
US11904081B2 (en) 2018-05-11 2024-02-20 Lupagen, Inc. Systems and methods for closed loop, real-time modifications of patient cells
WO2019226650A1 (en) 2018-05-23 2019-11-28 Modernatx, Inc. Delivery of dna
EP3581201A1 (de) 2018-06-15 2019-12-18 GlaxoSmithKline Biologicals S.A. Escherichia coli o157:h7 polypeptide und verwendungen davon
WO2020023390A1 (en) 2018-07-25 2020-01-30 Modernatx, Inc. Mrna based enzyme replacement therapy combined with a pharmacological chaperone for the treatment of lysosomal storage disorders
WO2020035609A2 (en) 2018-08-17 2020-02-20 Glaxosmithkline Biologicals Sa Immunogenic compositions and uses thereof
US20220110966A1 (en) 2018-09-02 2022-04-14 Modernatx, Inc. Polynucleotides encoding very long-chain acyl-coa dehydrogenase for the treatment of very long-chain acyl-coa dehydrogenase deficiency
US20230009009A1 (en) 2018-09-13 2023-01-12 Modernatx, Inc. Polynucleotides encoding glucose-6-phosphatase for the treatment of glycogen storage disease
US20220243182A1 (en) 2018-09-13 2022-08-04 Modernatx, Inc. Polynucleotides encoding branched-chain alpha-ketoacid dehydrogenase complex e1-alpha, e1-beta, and e2 subunits for the treatment of maple syrup urine disease
AU2019339430A1 (en) 2018-09-14 2021-04-29 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide A1 for the treatment of Crigler-Najjar Syndrome
WO2020069169A1 (en) 2018-09-27 2020-04-02 Modernatx, Inc. Polynucleotides encoding arginase 1 for the treatment of arginase deficiency
TW202043256A (zh) 2019-01-10 2020-12-01 美商健生生物科技公司 前列腺新抗原及其用途
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
BR112021017637A8 (pt) 2019-03-08 2022-08-16 Massachusetts Inst Technology Vírus oncolítico sintético, composição farmacêutica, e, método para tratar câncer
CN114375190A (zh) 2019-05-08 2022-04-19 阿斯利康(瑞典)有限公司 用于皮肤和伤口的组合物及其使用方法
CA3140019A1 (en) 2019-05-30 2020-12-03 Gritstone Bio, Inc. Modified adenoviruses
WO2020255011A1 (en) 2019-06-18 2020-12-24 Janssen Sciences Ireland Unlimited Company Combination of hepatitis b virus (hbv) vaccines and anti-pd-1 or anti-pd-l1 antibody
WO2020255010A1 (en) 2019-06-18 2020-12-24 Janssen Sciences Ireland Unlimited Company Combination of recombinant interleukin 12 construct and hepatitis b virus (hbv) vaccines
WO2020255009A2 (en) 2019-06-18 2020-12-24 Janssen Sciences Ireland Unlimited Company Combination of hepatitis b virus (hbv) vaccines and anti-pd-1 antibody
CN114340664A (zh) 2019-06-18 2022-04-12 爱尔兰詹森科学公司 乙型肝炎病毒(HBV)疫苗和靶向HBV的RNAi的组合
EP3986915A1 (de) 2019-06-18 2022-04-27 Janssen Sciences Ireland Unlimited Company Konstrukt aus rekombinantem interleukin 12 und verwendungen davon
MA56536A (fr) 2019-06-20 2022-04-27 Janssen Sciences Ireland Unlimited Co Administration de nanoparticules lipidiques ou de liposomes de vaccins contre le virus de l'hépatite b (vhb)
US20220273789A1 (en) 2019-07-21 2022-09-01 Glaxosmithkline Biologicals Sa Therapeutic viral vaccine
MX2022003269A (es) 2019-09-19 2022-07-04 Modernatx Inc Compuestos lipidicos de cola ramificada y composiciones para la administracion intracelular de agentes terapeuticos.
WO2021099906A1 (en) 2019-11-18 2021-05-27 Janssen Biotech, Inc. Vaccines based on mutant calr and jak2 and their uses
JP2023517644A (ja) 2020-03-09 2023-04-26 アークトゥラス・セラピューティクス・インコーポレイテッド コロナウイルスワクチン組成物及び方法
WO2021209970A1 (en) 2020-04-16 2021-10-21 Glaxosmithkline Biologicals Sa Sars cov-2 spike protein construct
JP2023527875A (ja) 2020-06-01 2023-06-30 モダーナティエックス・インコーポレイテッド フェニルアラニンヒドロキシラーゼバリアント及びその使用
WO2021245611A1 (en) 2020-06-05 2021-12-09 Glaxosmithkline Biologicals Sa Modified betacoronavirus spike proteins
WO2022002783A1 (en) 2020-06-29 2022-01-06 Glaxosmithkline Biologicals Sa Adjuvants
TW202216190A (zh) 2020-07-08 2022-05-01 愛爾蘭商健生科學愛爾蘭無限公司 抗hbv之rna複製子疫苗
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
US20230406895A1 (en) 2020-11-13 2023-12-21 Modernatx, Inc. Polynucleotides encoding cystic fibrosis transmembrane conductance regulator for the treatment of cystic fibrosis
WO2022133230A1 (en) 2020-12-18 2022-06-23 Janssen Pharmaceuticals, Inc. Combination therapy for treating hepatitis b virus infection
WO2022135993A2 (en) * 2020-12-22 2022-06-30 Curevac Ag Pharmaceutical composition comprising lipid-based carriers encapsulating rna for multidose administration
WO2022137128A2 (en) 2020-12-23 2022-06-30 Glaxosmithkline Biologicals Sa Self-amplifying messenger rna
CA3203442A1 (en) 2020-12-28 2022-07-07 Arcturus Therapeutics, Inc. Transcription activator-like effector nucleases (talens) targeting hbv
EP4032546A1 (de) 2021-01-20 2022-07-27 GlaxoSmithKline Biologicals S.A. Therapeutischer viraler impfstoff
US11524023B2 (en) 2021-02-19 2022-12-13 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase and uses thereof
US20240189449A1 (en) 2021-03-24 2024-06-13 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding ornithine transcarbamylase for the treatment of ornithine transcarbamylase deficiency
US20240207374A1 (en) 2021-03-24 2024-06-27 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding glucose-6-phosphatase and uses thereof
WO2022204369A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
US20240216288A1 (en) 2021-03-24 2024-07-04 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits and uses thereof
US20240181037A1 (en) 2021-03-26 2024-06-06 Glaxosmithkline Biologicals Sa Immunogenic compositions
EP4346894A1 (de) 2021-05-24 2024-04-10 GlaxoSmithKline Biologicals S.A. Adjuvans
EP4352247A1 (de) 2021-06-09 2024-04-17 GlaxoSmithKline Biologicals s.a. Freisetzungstest zur bestimmung der wirksamkeit eines selbstverstärkenden rna-wirkstoffprodukts und verfahren zur verwendung
EP4355882A2 (de) 2021-06-15 2024-04-24 Modernatx, Inc. Manipulierte polynukleotide zur zelltyp- oder mikroumgebungsspezifischen expression
WO2022271776A1 (en) 2021-06-22 2022-12-29 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2023020992A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Novel methods
WO2023020993A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Novel methods
WO2023020994A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Novel methods
WO2023021427A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Freeze-drying of lipid nanoparticles (lnps) encapsulating rna and formulations thereof
EP4387596A1 (de) 2021-08-16 2024-06-26 GlaxoSmithKline Biologicals SA Lyophilisierte rna-impfstoffe mit niedriger dosis und verfahren zur herstellung und verwendung davon
CN118043466A (zh) 2021-09-03 2024-05-14 葛兰素史克生物有限公司 自扩增信使核糖核酸中核苷酸碱基的取代
WO2023056044A1 (en) 2021-10-01 2023-04-06 Modernatx, Inc. Polynucleotides encoding relaxin for the treatment of fibrosis and/or cardiovascular disease
CN115960922A (zh) * 2021-10-09 2023-04-14 吴可行 自复制rna分子设计及其应用
AU2022386716A1 (en) 2021-11-15 2024-05-02 Technische Universität Dresden Site-specific recombinases for efficient and specific genome editing
WO2023183909A2 (en) 2022-03-25 2023-09-28 Modernatx, Inc. Polynucleotides encoding fanconi anemia, complementation group proteins for the treatment of fanconi anemia
WO2023198815A1 (en) 2022-04-14 2023-10-19 Janssen Vaccines & Prevention B.V. Sequential administration of adenoviruses
WO2023218420A1 (en) 2022-05-13 2023-11-16 Janssen Pharmaceuticals, Inc. Mrna compositions for inducing latent hiv-1 reversal
CN117070464A (zh) * 2022-05-16 2023-11-17 上海行深生物科技有限公司 蛋白包裹自复制rna及其制备方法
WO2023233290A1 (en) 2022-05-31 2023-12-07 Janssen Sciences Ireland Unlimited Company Rnai agents targeting pd-l1
WO2023242817A2 (en) 2022-06-18 2023-12-21 Glaxosmithkline Biologicals Sa Recombinant rna molecules comprising untranslated regions or segments encoding spike protein from the omicron strain of severe acute respiratory coronavirus-2
WO2024026254A1 (en) 2022-07-26 2024-02-01 Modernatx, Inc. Engineered polynucleotides for temporal control of expression
WO2024068545A1 (en) 2022-09-26 2024-04-04 Glaxosmithkline Biologicals Sa Influenza virus vaccines
WO2024133160A1 (en) 2022-12-19 2024-06-27 Glaxosmithkline Biologicals Sa Hepatitis b compositions
GB202404607D0 (en) 2024-03-29 2024-05-15 Glaxosmithkline Biologicals Sa RNA formulation

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000000617A2 (en) 1998-06-29 2000-01-06 U.S. Medical Research Institute Of Infectious Diseases Ebola virion proteins expressed from venezuelan equine encephalitis (vee) virus replicons
US6090406A (en) 1984-04-12 2000-07-18 The Liposome Company, Inc. Potentiation of immune responses with liposomal adjuvants
WO2001029233A2 (en) 1999-10-20 2001-04-26 The Johns Hopkins University School Of Medicine Chimeric immunogenic compositions and nucleic acids encoding them
WO2002009645A2 (en) 2000-08-01 2002-02-07 The Johns Hopkins University Intercellular transport protein linked to an antigen as a molecular vaccine
WO2002026209A2 (en) 2000-09-28 2002-04-04 Chiron Corporation Microparticles for delivery of the heterologous nucleic acids
WO2002061113A2 (en) 2001-02-01 2002-08-08 The Johns Hopkins University Nucleic acid derived vaccine that encodes an antigen linked to a polypeptide that promotes antigen presentation
WO2002072027A2 (en) 2001-03-14 2002-09-19 University Of Alabama Research Foundation Oncolytic rna replicons
WO2002079239A2 (en) 2001-01-31 2002-10-10 U.S. Army Medical Research Institute Of Infectious Diseases Chimeric filovirus glycoprotein
WO2002095023A2 (en) 2001-05-23 2002-11-28 Institut Pasteur Replicons derived from positive strand rna virus genomes useful for the production of heterologous proteins
WO2006078294A2 (en) 2004-05-21 2006-07-27 Novartis Vaccines And Diagnostics Inc. Alphavirus vectors for respiratory pathogen vaccines
WO2007047749A1 (en) 2005-10-18 2007-04-26 Novartis Vaccines And Diagnostics Inc. Mucosal and systemic immunizations with alphavirus replicon particles
WO2009146867A1 (en) 2008-06-04 2009-12-10 Institut Für Viruskrankheiten Und Immunprophylaxe Pestivirus replicons providing an rna-based viral vector system
WO2012006378A1 (en) 2010-07-06 2012-01-12 Novartis Ag Liposomes with lipids having an advantageous pka- value for rna delivery
WO2012030901A1 (en) 2010-08-31 2012-03-08 Novartis Ag Small liposomes for delivery of immunogen-encoding rna
EP2591114B1 (de) 2010-07-06 2016-06-08 GlaxoSmithKline Biologicals SA Immunisierung grosser säugetiere durch niedrige dosen von rna

Family Cites Families (100)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5140086A (en) 1988-11-25 1992-08-18 Weyerhaeuser Company Isocyanate modified cellulose products and method for their manufacture
US6867195B1 (en) 1989-03-21 2005-03-15 Vical Incorporated Lipid-mediated polynucleotide administration to reduce likelihood of subject's becoming infected
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5279833A (en) 1990-04-04 1994-01-18 Yale University Liposomal transfection of nucleic acids into animal cells
US5693535A (en) 1992-05-14 1997-12-02 Ribozyme Pharmaceuticals, Inc. HIV targeted ribozymes
US5750390A (en) 1992-08-26 1998-05-12 Ribozyme Pharmaceuticals, Inc. Method and reagent for treatment of diseases caused by expression of the bcl-2 gene
IL105914A0 (en) 1992-06-04 1993-10-20 Univ California Methods and compositions for in vivo gene therapy
JPH07509133A (ja) 1992-07-17 1995-10-12 リボザイム・ファーマシューティカルズ・インコーポレイテッド 動物疾患の処置のための方法および剤
EP1624068A1 (de) 1993-06-01 2006-02-08 Life Technologies Inc. Genetische Impfung mit kationischen Lipidstoffen
US6015686A (en) 1993-09-15 2000-01-18 Chiron Viagene, Inc. Eukaryotic layered vector initiation systems
AU727923B2 (en) * 1995-09-27 2001-01-04 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Production of infectious respiratory syncytial virus from cloned nucleotide sequences
WO1998000110A1 (en) 1996-07-03 1998-01-08 University Of Pittsburgh Emulsion formulations for hydrophilic active agents
EP0938298B1 (de) 1996-09-13 2002-12-04 Lipoxen Technologies Limited Liposomenzusammensetzung
US7384923B2 (en) 1999-05-14 2008-06-10 Lipoxen Technologies Limited Liposomes
US6395302B1 (en) 1996-11-19 2002-05-28 Octoplus B.V. Method for the preparation of microspheres which contain colloidal systems
EP1027033B1 (de) 1997-05-14 2009-07-22 The University Of British Columbia Hochwirksame verkapselung von nukleinsäuren in lipidvesikeln
US6060308A (en) 1997-09-04 2000-05-09 Connaught Laboratories Limited RNA respiratory syncytial virus vaccines
US6009406A (en) 1997-12-05 1999-12-28 Square D Company Methodology and computer-based tools for re-engineering a custom-engineered product line
US6432925B1 (en) 1998-04-16 2002-08-13 John Wayne Cancer Institute RNA cancer vaccine and methods for its use
DE69923840T2 (de) 1999-09-09 2006-04-06 Curevac Gmbh Transfer von mRNAs unter Verwendung von polykationischen Verbindungen
US8541008B2 (en) * 1999-11-19 2013-09-24 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Pharmaceutical compositions and methods to vaccinate against candidiasis
US20030212022A1 (en) * 2001-03-23 2003-11-13 Jean-Marie Vogel Compositions and methods for gene therapy
US7149665B2 (en) 2000-04-03 2006-12-12 Browzwear International Ltd System and method for simulation of virtual wear articles on virtual models
WO2001079253A1 (en) 2000-04-18 2001-10-25 Human Genome Sciences, Inc. Extracellular matrix polynucleotides, polypeptides, and antibodies
AU2001276619A1 (en) 2000-07-03 2002-01-14 Chiron S.P.A. Immunisation against chlamydia pneumoniae
EP2189473A3 (de) 2000-10-27 2010-08-11 Novartis Vaccines and Diagnostics S.r.l. Nukleinsäure und Proteine von Gruppen A und B Streptokokken
DE50214200D1 (de) 2001-06-05 2010-03-25 Curevac Gmbh Stabilisierte Tumorantigen-mRNA mit erhöhtem G/C-Gehalt
EP1423142A1 (de) 2001-08-31 2004-06-02 Chiron SRL. Helicobacter pylori impfung mit einer kombination der proteine caga, vaca und nap
DE60233061D1 (de) * 2001-09-06 2009-09-03 Alphavax Inc Alphavirus replikon-vektorsysteme
US20050163832A1 (en) 2002-02-13 2005-07-28 Vladimir Torchilin Intracellular delivery of therapeutic agents
DE10207177A1 (de) 2002-02-19 2003-09-04 Novosom Ag Fakultativ kationische Lipide
US7901708B2 (en) 2002-06-28 2011-03-08 Protiva Biotherapeutics, Inc. Liposomal apparatus and manufacturing methods
WO2004024919A1 (en) 2002-09-13 2004-03-25 Replicor, Inc. Non-sequence complementary antiviral oligonucleotides
EP1587816B1 (de) 2002-12-23 2010-06-16 Vical Incorporated Impfstoffe gegen infektionen mit dem humanen zytomegalivirus auf grundlage von codonoptimierten polynukleotiden
US8338583B2 (en) 2003-02-04 2012-12-25 Bar-Ilan University Snornai-small nucleolar RNA degradation by RNA interference in trypanosomatids
US7731967B2 (en) 2003-04-30 2010-06-08 Novartis Vaccines And Diagnostics, Inc. Compositions for inducing immune responses
EP1637144A4 (de) 2003-05-30 2010-01-13 Nippon Shinyaku Co Ltd Oligonucleinsäure enthaltender komposit und diesen enthaltende pharmazeutische zusammensetzung
US20100255002A1 (en) 2003-06-26 2010-10-07 Chiron Corporation Immunogenic compositions for chlamydia trachomatis
US7368537B2 (en) * 2003-07-15 2008-05-06 Id Biomedical Corporation Of Quebec Subunit vaccine against respiratory syncytial virus infection
EP1648519B1 (de) 2003-07-16 2014-10-08 Protiva Biotherapeutics Inc. Lipid verkapselte interferierende rns
ES2505695T3 (es) 2003-07-31 2014-10-10 Novartis Vaccines And Diagnostics, Inc. Composiciones inmunógenas para Streptococcus pyogenes
US7303881B2 (en) 2004-04-30 2007-12-04 Pds Biotechnology Corporation Antigen delivery compositions and methods of use
GB0410866D0 (en) 2004-05-14 2004-06-16 Chiron Srl Haemophilius influenzae
US20060024670A1 (en) 2004-05-18 2006-02-02 Luke Catherine J Influenza virus vaccine composition and methods of use
ATE420965T1 (de) 2004-05-18 2009-01-15 Alphavax Inc Von tc-83 abgeleitete alphavirus-vektoren, partikel und verfahren
AU2005252273B2 (en) 2004-06-07 2011-04-28 Arbutus Biopharma Corporation Lipid encapsulated interfering RNA
AU2005251403B2 (en) 2004-06-07 2011-09-01 Arbutus Biopharma Corporation Cationic lipids and methods of use
WO2006007712A1 (en) * 2004-07-19 2006-01-26 Protiva Biotherapeutics, Inc. Methods comprising polyethylene glycol-lipid conjugates for delivery of therapeutic agents
JP5086082B2 (ja) 2004-10-01 2012-11-28 ノバルティス ヴァクシンズ アンド ダイアグノスティクス エスアールエル C型肝炎ウイルス複製系
WO2007013893A2 (en) * 2004-11-15 2007-02-01 Novartis Vaccines And Diagnostics Inc. Immunogenic compositions containing anthrax antigen, biodegradable polymer microparticles, and polynucleotide-containing immunological adjuvant
MX291624B (es) 2005-02-18 2011-11-04 Novartis Vaccines & Diagnostic Inmunogenos de escherichia coli uropatogenica.
AU2006214064B2 (en) 2005-02-18 2012-04-26 J. Craig Venter Institute, Inc. Proteins and nucleic acids from meningitis/sepsis-associated escherichia coli
US20080138431A1 (en) 2005-03-02 2008-06-12 James Edward Eyles Pharmaceutical Composition
AU2006235045A1 (en) 2005-03-30 2006-10-19 J. Craig Venter Institute, Inc. Haemophilus influenzae type b
US7618393B2 (en) 2005-05-03 2009-11-17 Pharmajet, Inc. Needle-less injector and method of fluid delivery
JP2008544745A (ja) 2005-05-12 2008-12-11 ノバルティス ヴァクシンズ アンド ダイアグノスティクス, インコーポレイテッド Chlamydiatrachomatisのための免疫原性組成物
US8703095B2 (en) 2005-07-07 2014-04-22 Sanofi Pasteur S.A. Immuno-adjuvant emulsion
ES2735531T3 (es) 2005-08-23 2019-12-19 Univ Pennsylvania ARN que contiene nucleósidos modificados y métodos de uso del mismo
JP2007112768A (ja) 2005-10-24 2007-05-10 Kyoto Univ 肝指向性リポソーム組成物
EP1954306A2 (de) 2005-10-25 2008-08-13 Novartis Vaccines and Diagnostics S.r.l. Zusammensetzungen mit yersinia pestis-antigenen
US7915399B2 (en) 2006-06-09 2011-03-29 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
JP2010500399A (ja) 2006-08-16 2010-01-07 ノバルティス アーゲー 尿路病原性大腸菌由来の免疫原
JP2010519203A (ja) 2007-02-16 2010-06-03 メルク・シャープ・エンド・ドーム・コーポレイション 生物活性分子の活性を強化するための組成物及び方法
US8877206B2 (en) 2007-03-22 2014-11-04 Pds Biotechnology Corporation Stimulation of an immune response by cationic lipids
DK2494993T3 (en) 2007-05-04 2018-11-12 Marina Biotech Inc Amino acid lipids and uses thereof
DE102007029471A1 (de) 2007-06-20 2008-12-24 Novosom Ag Neue fakultativ kationische Sterole
GB0714963D0 (en) 2007-08-01 2007-09-12 Novartis Ag Compositions comprising antigens
GB0717187D0 (en) 2007-09-04 2007-10-17 Novartis Ag Compositions comprising yersinia pestis antigens
WO2009042794A2 (en) 2007-09-26 2009-04-02 Vanderbilt University Venezuelan equine encephalitis replicons expressing paramyxovirus glycoproteins as vaccine
US9234181B2 (en) 2007-11-26 2016-01-12 Novartis Ag RNA expression cassette and cells for making alphavirus particles
WO2009111088A2 (en) 2008-01-02 2009-09-11 The Johns Hopkins University Antitumor immunization by liposomal delivery of vaccine to the spleen
CA3044134A1 (en) 2008-01-02 2009-07-09 Arbutus Biopharma Corporation Improved compositions and methods for the delivery of nucleic acids
ITMI20081249A1 (it) 2008-07-09 2010-01-09 Novartis Vaccines & Diagnostic Immunogeni di escherichia coli con solubilità migliorata.
WO2009109860A2 (en) 2008-03-06 2009-09-11 Novartis Ag Mutant forms of chlamydia htra
CN102119217B (zh) 2008-04-15 2015-06-03 普洛体维生物治疗公司 用于核酸递送的新型制剂
WO2009127230A1 (en) 2008-04-16 2009-10-22 Curevac Gmbh MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION
WO2009132206A1 (en) 2008-04-25 2009-10-29 Liquidia Technologies, Inc. Compositions and methods for intracellular delivery and release of cargo
US20100040650A1 (en) * 2008-05-30 2010-02-18 Crowe Jr James E Virus-Like paramyxovirus particles and vaccines
CL2008002322A1 (es) 2008-08-07 2009-06-05 Univ Concepcion Formulacion farmaceutica veterinaria que comprende un sistema vectorial viral constituido por una particula recombinante de arn que codifica una cu/zn superoxido dismutasa de la bacteria patogena de bovinos brucella abortus, y al menos un alfavirus arn perteneciente a la familia del virus semliki forest (sfv), util como vacuna.
CA2740000C (en) 2008-10-09 2017-12-12 Tekmira Pharmaceuticals Corporation Improved amino lipids and methods for the delivery of nucleic acids
JP6046348B2 (ja) 2008-11-18 2016-12-14 タケダ ヴァクシーンズ, インコーポレイテッド Rsvfvlpならびにその製造および使用の方法
JP5830009B2 (ja) 2009-04-14 2015-12-09 ノバルティス アーゲー Staphylococcusaureusに対して免疫化するための組成物
CA2767127A1 (en) 2009-07-01 2011-01-06 Protiva Biotherapeutics, Inc. Novel lipid formulations for delivery of therapeutic agents to solid tumors
EP2450031B1 (de) 2009-07-02 2018-08-29 Konica Minolta Holdings, Inc. Verfahren zur herstellung von liposomen durch ein zweistufen-emulgierverfahren unter verwendung der äusseren wässrigen phase mit spezifischen dispergiermitteln, verfahren zur herstellung der liposom-dispersion oder trockenpulver durch verwendung des verfahrens zur herstellung von liposomen und liposomdispersion oder trockenpulver daraus
CA2766907A1 (en) 2009-07-06 2011-01-13 Novartis Ag Self replicating rna molecules and uses thereof
US20120164176A1 (en) 2009-07-15 2012-06-28 Kurt Swanson Rsv f protein compositions amd methods for making same
US8277919B2 (en) 2009-07-23 2012-10-02 VMO Systems, Inc. Reflective coating for an optical disc
DK2459231T3 (en) 2009-07-31 2016-09-05 Ethris Gmbh RNA with a combination of unmodified and modified nucleotides for protein expression
CN107028886A (zh) * 2009-11-04 2017-08-11 不列颠哥伦比亚大学 含有核酸的脂质粒子及相关的方法
US20110112353A1 (en) 2009-11-09 2011-05-12 Circulite, Inc. Bifurcated outflow cannulae
NZ700688A (en) 2009-12-01 2016-02-26 Shire Human Genetic Therapies Delivery of mrna for the augmentation of proteins and enzymes in human genetic diseases
AU2010334911A1 (en) 2009-12-23 2012-07-12 Novartis Ag Lipids, lipid compositions, and methods of using them
RS63817B1 (sr) 2010-07-06 2023-01-31 Glaxosmithkline Biologicals Sa Čestice za isporuku slične virionu za molekule samoreplicirajuće rnk
US9770463B2 (en) 2010-07-06 2017-09-26 Glaxosmithkline Biologicals Sa Delivery of RNA to different cell types
DK3243526T3 (da) 2010-07-06 2020-02-17 Glaxosmithkline Biologicals Sa Levering af rna til at udløse flere immunsignalveje
JP6025721B2 (ja) 2010-07-06 2016-11-16 ノバルティス アーゲー カチオン性水中油型エマルジョン
BR112013004865A2 (pt) 2010-08-31 2016-06-07 Novartis Ag lípidos adequados para entrega lipossomal de codificadores de proteínas rna
EP4119155A1 (de) 2010-08-31 2023-01-18 GlaxoSmithKline Biologicals S.A. Pegylierte liposomen zur auslieferung von immunogen-kodierende rns
JP2014520806A (ja) 2011-07-06 2014-08-25 ノバルティス アーゲー Rna分子の送達のための有用なn:p比を有するリポソーム
MX366055B (es) 2011-08-31 2019-06-26 Novartis Ag Liposomas pegilados para admistracion de acido ribonucleico (arn) que codifica para inmunogeno.

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6090406A (en) 1984-04-12 2000-07-18 The Liposome Company, Inc. Potentiation of immune responses with liposomal adjuvants
WO2000000617A2 (en) 1998-06-29 2000-01-06 U.S. Medical Research Institute Of Infectious Diseases Ebola virion proteins expressed from venezuelan equine encephalitis (vee) virus replicons
WO2001029233A2 (en) 1999-10-20 2001-04-26 The Johns Hopkins University School Of Medicine Chimeric immunogenic compositions and nucleic acids encoding them
WO2002009645A2 (en) 2000-08-01 2002-02-07 The Johns Hopkins University Intercellular transport protein linked to an antigen as a molecular vaccine
WO2002026209A2 (en) 2000-09-28 2002-04-04 Chiron Corporation Microparticles for delivery of the heterologous nucleic acids
WO2002079239A2 (en) 2001-01-31 2002-10-10 U.S. Army Medical Research Institute Of Infectious Diseases Chimeric filovirus glycoprotein
WO2002061113A2 (en) 2001-02-01 2002-08-08 The Johns Hopkins University Nucleic acid derived vaccine that encodes an antigen linked to a polypeptide that promotes antigen presentation
WO2002072027A2 (en) 2001-03-14 2002-09-19 University Of Alabama Research Foundation Oncolytic rna replicons
WO2002095023A2 (en) 2001-05-23 2002-11-28 Institut Pasteur Replicons derived from positive strand rna virus genomes useful for the production of heterologous proteins
WO2006078294A2 (en) 2004-05-21 2006-07-27 Novartis Vaccines And Diagnostics Inc. Alphavirus vectors for respiratory pathogen vaccines
WO2007047749A1 (en) 2005-10-18 2007-04-26 Novartis Vaccines And Diagnostics Inc. Mucosal and systemic immunizations with alphavirus replicon particles
WO2009146867A1 (en) 2008-06-04 2009-12-10 Institut Für Viruskrankheiten Und Immunprophylaxe Pestivirus replicons providing an rna-based viral vector system
WO2012006378A1 (en) 2010-07-06 2012-01-12 Novartis Ag Liposomes with lipids having an advantageous pka- value for rna delivery
EP2591114B1 (de) 2010-07-06 2016-06-08 GlaxoSmithKline Biologicals SA Immunisierung grosser säugetiere durch niedrige dosen von rna
WO2012030901A1 (en) 2010-08-31 2012-03-08 Novartis Ag Small liposomes for delivery of immunogen-encoding rna

Non-Patent Citations (34)

* Cited by examiner, † Cited by third party
Title
ABERLE ET AL.: "Humoral and Cellular Immune Response to RNA Immunization with Flavivirus Replicons Derived from Tick-Borne Encephalitis Virus", JOURNAL OF VIROLOGY, vol. 79, no. 24, December 2005 (2005-12-01), pages 15107 - 15113, XP055361245
BRINGMANN ET AL.: "RNA Vaccines in Cancer Treatment';", J BIOMEDICINE AND BIOTECHNOLOGY, 2010, pages 1 - 12, XP002765542
BUYENS ET AL.: "Elucidating the Encapsulation of Short Interfering RNA in PEGylated Cationic Liposomes", LANGMUIR, vol. 25, no. 9, 2009, pages 4886 - 4891, XP055384662
CROOKE: "Antisense Drug Technology", 2007, article MACLACHLAN I: "chapter 9 Liposomal Formulations for Nucleic Acid Delivery", pages: 237 - 270, XP002584305
DAVID M. KNIPE, PETER M. HOWLEY: "Fields Virology, 4th edition", vol. 1, 2001, LIPINCOTT WILLIAMS & WILKINS, pages: 2pp, 690, XP055384675
FENSKE ET AL.: "Liposomal Nanomedicines: An Emerging Field", TOXICOLOGIC PATHOLOGY, vol. 36, no. 1, 2008, pages 21 - 29, XP055179556
FLEETON ET AL.: "Self-Replicative RNA Vaccines Elicit Protection against Influenza A Virus, Respiratory Syncytial Virus, and a Tickborne Encephalitis Virus", JOURNAL OF INFECTIOUS DISEASES, vol. 183, no. 9, May 2001 (2001-05-01), pages 1395 - 1398, XP002224503
GEALL ET AL.: "Nonviral delivery of self-amplifying RNA vaccines", PNAS, vol. 109, no. 36, 4 September 2012 (2012-09-04), pages 14604 - 14609, XP002683929
GEISBERT ET AL.: "Postexposure Protection of Guinea Pigs against a Lethal Ebola Virus Challenge Is Conferred by RNA Interference", JOURNAL OF INFECTIOUS DISEASES, vol. 193, no. 12, 1 January 2006 (2006-01-01), pages 1650 - 1657, XP008078155
HEINZ FRAENKEL-CONRAT, PAUL C. KIMBALL, JAY A. LEVY: "Virology second edition", 1988, PRENTICE- HALL INC., article "Togaviridae", pages: 2pp, 99, XP055384685
HEYES ET AL.: "Cationic lipid saturation influences intracellular delivery of encapsulated nucleic acids", J CONTROL RELEASE, vol. 107, no. 2, 3 October 2005 (2005-10-03), pages 276 - 287, XP005076220
HEYES ET AL.: "Cationic lipid saturation influences intracellular delivery of encapsulated nucleic acids", JOURNAL OF CONTROLLED RELEASE, vol. 107, no. 2, 3 October 2005 (2005-10-03), pages 276 - 287, XP005076220
JEFFS ET AL.: "A scalable, extrusion-free method for efficient liposomal encapsulation of plasmid DNA", PHARM RES, vol. 22, no. 3, 1 March 2005 (2005-03-01), pages 362 - 372, XP002422317
JEFFS ET AL.: "A Scalable, Extrusion-Free Method for Efficient Liposomal Encapsulation of Plasmid DNA", PHARMACEUTICAL RESEARCH, vol. 22, no. 3, March 2005 (2005-03-01), pages 362 - 372, XP002422317
KANEDA YASUFUMI: "New vector innovation for drug delivery: development of fusigenic non-viral particles", CURRENT DRUG TARGETS, BENTHAM SCIENCE PUBLISHER, US, vol. 4, no. 8, 1 November 2003 (2003-11-01), pages 599 - 602, XP009116733, ISSN: 1389-4501, DOI: 10.2174/1389450033490740 *
LUNDSTROM ET AL.: "Biology and application of alphaviruses in gene therapy", GENE THERAPY, vol. 12, 2005, pages S92 - S97
MANNING ET AL.: "Infectivity of Liposomally Encapsulated Nucleic Acids Isolated From EMC Virus and Scrapie-Infected Mouse Brain", INTERVIROLOGY, vol. 20, 1 January 1983 (1983-01-01), pages 164 - 168, XP001005390
MANNING ET AL.: "Infectivity of Liposomally Encapsulated Nucleic Acids Isolated From EMC Virus and Scrapie-Infected Mouse Brain", INTERVIROLOGY, vol. 20, 1983, pages 164 - 168, XP001005390
MARTINON ET AL.: "Induction of virus-specific cytotoxic T lymphocytes in vivo by liposome-entrapped mRNA", EUROPEAN JOURNAL OF IMMUNOLOGY, vol. 23, 1993, pages 1719 - 1722, XP000618955
MARTINON ET AL.: "Induction of virus-specific cytotoxic T lymphocytes in vivo by liposome-entrapped mRNA", EUROPEAN JOURNAL OF IMMUNOLOGY, vol. 23, no. 7, 1 January 1993 (1993-01-01), pages 1719 - 1722, XP000618955
OBATA Y; TAJIMA S; TAKEOKA S: "Evaluation of pH-responsive liposomes containing amino acid- based zwitterionic lipids for improving intracellular drug delivery in vitro and in vivo", JOURNAL OF CONTROLLED RELEASE, vol. 142, no. 2, 3 March 2010 (2010-03-03), pages 267 - 276, XP026905290
RAYNER ET AL.: "Alphavirus vectors and vaccination", REVIEWS IN MEDICAL VIROLOGY, vol. 12, no. 5, 1 September 2002 (2002-09-01), pages 279 - 296, XP008035758
RAYNER ET AL.: "Alphavirus vectors and vaccination", REVIEWS IN MEDICAL VIROLOGY, vol. 12, no. 5, 2002, pages 279 - 296, XP008035758
REN ET AL.: "Immunogene therapy of recurrent glioblastoma multiforme with a liposomally encapsulated replication-incompetent Semliki forest virus vector carrying the human interleukin-12 gene - a phase I/II clinical protocol", JOURNAL OF NEURO- ONCOLOGY, vol. 64, 1 January 2003 (2003-01-01), pages 147 - 154, XP055249047
SAXENA ET AL.: "Induction of immune responses and protection in mice against rabies using a self-replicating RNA vaccine encoding rabies virus glycoprotein", VETERINARY MICROBIOLOGY, vol. 136, 14 April 2009 (2009-04-14), pages 36 - 44, XP026071306
SAXENA ET AL.: "Induction of immune responses and protection in mice against rabies using a self-replicating RNA vaccine encoding rabies virus glycoprotein", VETERINARY MICROBIOLOGY, vol. 139, no. 1-2, 14 April 2009 (2009-04-14), pages 36 - 44, XP026071306
SCHIRRMACHER ET AL.: "Intra-pinna anti-tumor vaccination with self-replicating infectious RNA or with DNA encoding a model tumor antigen and a cytokine", GENE THERAPY, vol. 7, no. 13, 1 July 2000 (2000-07-01), pages 1137 - 1146, XP002359468
STREJAN ET AL.: "Suppression of Chronic-Relapsing Experimental Allergic Encephalomyelitis in Strain-13 Guinea Pigs by Administration of Liposome-Associated Myelin Basic Protein", JOURNAL OF NEUROIMMUNOLOGY, vol. 7, 1984, pages 27 - 41, XP025906071
VIGNUZZI ET AL.: "Naked RNA immunization with replicons derived from poliovirus and Semliki Forest virus genomes for the generation of a cytotoxic T cell response against the influenza A virus nucleoprotein", JOURNAL OF GENERAL VIROLOGY, vol. 82, no. 7, July 2001 (2001-07-01), pages 1737 - 1747, XP002224507
WANG ET AL.: "pH-sensitive immunoliposomes mediate target- cell -specific delivery and controlled expression of a foreign gene in mouse", PROC. NAT. ACAD. SCI. USA, vol. 84, November 1987 (1987-11-01), pages 7851 - 7855, XP002089781
WHITEHEAD ET AL.: "Knocking down barriers: advances in siRNA delivery", NATURE REVIEWS DRUG DISCOVERY, vol. 8, no. 2, 1 February 2009 (2009-02-01), pages 129 - 138, XP002559384
WILSON ET AL.: "The Introduction of Poliovirus RNA into Cells via Lipid Vesicles (Liposomes)", CELL, vol. 17, May 1979 (1979-05-01), pages 77 - 84, XP023875372
WILSON TAZEWELL, DEMETRIOS PAPAHADJOPOULOS, ROBERT TABER: "Biological properties of poliovirus encapsulated in lipid vesicles: Antibody resistance and infectivity in virus-resistant cells", PROC. NATL. ACAD. SCI. USA, vol. 74, no. 8, August 1977 (1977-08-01), pages 3471 - 3475, XP055384651
ZIMMERMANN ET AL.: "RNAi-mediated gene silencing in non-human primates", NATURE, vol. 441, no. 7089, 4 May 2006 (2006-05-04), pages 111 - 114, XP002412249

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11845925B2 (en) 2010-07-06 2023-12-19 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11913001B2 (en) 2010-07-06 2024-02-27 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11766401B2 (en) 2010-07-06 2023-09-26 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with immunogens
US11666534B2 (en) 2010-07-06 2023-06-06 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with viral immunogens
EP4005592B1 (de) 2010-07-06 2022-10-12 GlaxoSmithKline Biologicals S.A. Virionabgabepartikel für selbstreplizierende rna-moleküle
US11850305B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Method of making lipid formulations with RNA encoding immunogens
US11851660B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11638693B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising RNA encoding an immunogen and lipid formulations comprising mole percentage of lipids
US11638694B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising lipid formulations and RNA encoding multiple immunogens
US11291682B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11324770B2 (en) 2010-07-06 2022-05-10 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US20220125723A1 (en) 2010-07-06 2022-04-28 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US11291635B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biological Sa Virion-like delivery particles for self-replicating RNA molecules
US11786467B2 (en) 2010-07-06 2023-10-17 Glaxosmithkline Biologicals Sa Lipid formulations with immunogens
US11839686B2 (en) 2010-07-06 2023-12-12 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
EP2729126B1 (de) 2011-07-06 2020-12-23 GlaxoSmithKline Biologicals SA Liposomen mit nützlichem n:p-verhältnis zur freisetzung von rna-molekülen
US11510973B2 (en) 2017-05-08 2022-11-29 Gritstone Bio, Inc. Alphavirus antigen vectors
US11504421B2 (en) 2017-05-08 2022-11-22 Gritstone Bio, Inc. Alphavirus neoantigen vectors
US11771747B2 (en) 2020-08-06 2023-10-03 Gritstone Bio, Inc. Multiepitope vaccine cassettes

Also Published As

Publication number Publication date
SI2590676T1 (sl) 2016-11-30
HUE060788T2 (hu) 2023-04-28
HRP20161352T1 (hr) 2016-12-02
JP2013533747A (ja) 2013-08-29
SI4005592T1 (sl) 2023-03-31
JP6061849B2 (ja) 2017-01-18
ES2934240T3 (es) 2023-02-20
PL4005592T3 (pl) 2023-02-06
WO2012006376A3 (en) 2012-04-19
CN106421773A (zh) 2017-02-22
CY1118080T1 (el) 2017-06-28
BR112013000392A2 (pt) 2021-05-25
LT2590676T (lt) 2016-10-25
AU2011276232A1 (en) 2013-02-21
PL2590676T3 (pl) 2017-02-28
WO2012006376A2 (en) 2012-01-12
US11291635B2 (en) 2022-04-05
US20220192997A1 (en) 2022-06-23
PT4005592T (pt) 2022-12-30
RS63817B1 (sr) 2023-01-31
ES2600892T3 (es) 2017-02-13
BR112013000392B8 (pt) 2022-10-04
HUE031485T2 (en) 2017-07-28
SMT201600386B (it) 2017-01-10
CA2804494A1 (en) 2012-01-12
US20130195968A1 (en) 2013-08-01
FI4005592T3 (fi) 2023-01-13
DK2590676T3 (en) 2016-10-24
EP4005592A1 (de) 2022-06-01
LT4005592T (lt) 2023-01-10
BR112013000392B1 (pt) 2022-07-12
EP3115061A1 (de) 2017-01-11
CN103052400A (zh) 2013-04-17
HRP20221522T1 (hr) 2023-02-17
CN103052400B (zh) 2016-11-16
EP4005592B1 (de) 2022-10-12
EP2590676A2 (de) 2013-05-15
PT2590676T (pt) 2016-11-04
AU2011276232B2 (en) 2016-01-14
EP4180057A1 (de) 2023-05-17
RS55260B1 (sr) 2017-02-28
RU2013104890A (ru) 2014-08-20
MX2013000089A (es) 2013-02-27
MX342608B (es) 2016-10-06

Similar Documents

Publication Publication Date Title
US11883534B2 (en) Immunisation with lipid formulations with RNA encoding immunogens
EP2590676B1 (de) Virion-artige transferpartikel für selbstreplizierende rna-moleküle
AU2018204178B2 (en) Small liposomes for delivery of immunogen-encoding RNA
US20200113830A1 (en) Pegylated liposomes for delivery of immunogen-encoding rna
EP2729126B1 (de) Liposomen mit nützlichem n:p-verhältnis zur freisetzung von rna-molekülen

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20130131

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20140821

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20160307

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: GLAXOSMITHKLINE BIOLOGICALS SA

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 820414

Country of ref document: AT

Kind code of ref document: T

Effective date: 20160915

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: ISLER AND PEDRAZZINI AG, CH

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602011029342

Country of ref document: DE

REG Reference to a national code

Ref country code: HR

Ref legal event code: TUEP

Ref document number: P20161352

Country of ref document: HR

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20161020

REG Reference to a national code

Ref country code: RO

Ref legal event code: EPE

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Ref document number: 2590676

Country of ref document: PT

Date of ref document: 20161104

Kind code of ref document: T

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20161026

REG Reference to a national code

Ref country code: NO

Ref legal event code: T2

Effective date: 20160817

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: HR

Ref legal event code: T1PR

Ref document number: P20161352

Country of ref document: HR

REG Reference to a national code

Ref country code: EE

Ref legal event code: FG4A

Ref document number: E012605

Country of ref document: EE

Effective date: 20161101

REG Reference to a national code

Ref country code: SK

Ref legal event code: T3

Ref document number: E 22420

Country of ref document: SK

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2600892

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20170213

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20160402422

Country of ref document: GR

Effective date: 20170117

REG Reference to a national code

Ref country code: DE

Ref legal event code: R026

Ref document number: 602011029342

Country of ref document: DE

PLBI Opposition filed

Free format text: ORIGINAL CODE: 0009260

26 Opposition filed

Opponent name: BAYER INTELLECTUAL PROPERTY GMBH/BAYER PHARMA AKTI

Effective date: 20170515

Opponent name: MUELLER, CHRISTIAN

Effective date: 20170516

PLAX Notice of opposition and request to file observation + time limit sent

Free format text: ORIGINAL CODE: EPIDOSNOBS2

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 7

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E031485

Country of ref document: HU

PLAF Information modified related to communication of a notice of opposition and request to file observations + time limit

Free format text: ORIGINAL CODE: EPIDOSCOBS2

PLBB Reply of patent proprietor to notice(s) of opposition received

Free format text: ORIGINAL CODE: EPIDOSNOBS3

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 8

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 820414

Country of ref document: AT

Kind code of ref document: T

Effective date: 20160817

PLAB Opposition data, opponent's data or that of the opponent's representative modified

Free format text: ORIGINAL CODE: 0009299OPPO

R26 Opposition filed (corrected)

Opponent name: MUELLER, CHRISTIAN

Effective date: 20170516

APBM Appeal reference recorded

Free format text: ORIGINAL CODE: EPIDOSNREFNO

APBP Date of receipt of notice of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA2O

APAH Appeal reference modified

Free format text: ORIGINAL CODE: EPIDOSCREFNO

APBM Appeal reference recorded

Free format text: ORIGINAL CODE: EPIDOSNREFNO

APBP Date of receipt of notice of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA2O

APBM Appeal reference recorded

Free format text: ORIGINAL CODE: EPIDOSNREFNO

APBP Date of receipt of notice of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA2O

APAL Date of receipt of statement of grounds of an appeal modified

Free format text: ORIGINAL CODE: EPIDOSCNOA3O

APBQ Date of receipt of statement of grounds of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA3O

APBQ Date of receipt of statement of grounds of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA3O

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20161352

Country of ref document: HR

Payment date: 20190704

Year of fee payment: 9

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20161352

Country of ref document: HR

Payment date: 20200709

Year of fee payment: 10

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20200615

Year of fee payment: 10

Ref country code: RO

Payment date: 20200706

Year of fee payment: 10

REG Reference to a national code

Ref country code: DE

Ref legal event code: R103

Ref document number: 602011029342

Country of ref document: DE

Ref country code: DE

Ref legal event code: R064

Ref document number: 602011029342

Country of ref document: DE

APBU Appeal procedure closed

Free format text: ORIGINAL CODE: EPIDOSNNOA9O

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20161352

Country of ref document: HR

Payment date: 20210701

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SM

Payment date: 20210623

Year of fee payment: 11

Ref country code: PT

Payment date: 20210625

Year of fee payment: 11

Ref country code: SK

Payment date: 20210630

Year of fee payment: 11

Ref country code: FI

Payment date: 20210622

Year of fee payment: 11

Ref country code: EE

Payment date: 20210625

Year of fee payment: 11

Ref country code: LT

Payment date: 20210622

Year of fee payment: 11

Ref country code: IT

Payment date: 20210622

Year of fee payment: 11

Ref country code: MC

Payment date: 20210625

Year of fee payment: 11

Ref country code: NO

Payment date: 20210624

Year of fee payment: 11

Ref country code: LU

Payment date: 20210625

Year of fee payment: 11

Ref country code: NL

Payment date: 20210622

Year of fee payment: 11

Ref country code: CZ

Payment date: 20210628

Year of fee payment: 11

Ref country code: FR

Payment date: 20210622

Year of fee payment: 11

Ref country code: GR

Payment date: 20210624

Year of fee payment: 11

RDAF Communication despatched that patent is revoked

Free format text: ORIGINAL CODE: EPIDOSNREV1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: PATENT REVOKED

RDAG Patent revoked

Free format text: ORIGINAL CODE: 0009271

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: PATENT REVOKED

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: LT

Ref legal event code: MM9A

Effective date: 20310706

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20210623

Year of fee payment: 11

Ref country code: IE

Payment date: 20210624

Year of fee payment: 11

Ref country code: PL

Payment date: 20210624

Year of fee payment: 11

Ref country code: SE

Payment date: 20210623

Year of fee payment: 11

Ref country code: LV

Payment date: 20210622

Year of fee payment: 11

Ref country code: BE

Payment date: 20210622

Year of fee payment: 11

Ref country code: DK

Payment date: 20210624

Year of fee payment: 11

Ref country code: CH

Payment date: 20210622

Year of fee payment: 11

Ref country code: GB

Payment date: 20210623

Year of fee payment: 11

REG Reference to a national code

Ref country code: FI

Ref legal event code: MGE

27W Patent revoked

Effective date: 20210615

GBPR Gb: patent revoked under art. 102 of the ep convention designating the uk as contracting state

Effective date: 20210615

REG Reference to a national code

Ref country code: SK

Ref legal event code: MC4A

Ref document number: E 22420

Country of ref document: SK

Effective date: 20210615

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MK

Payment date: 20210623

Year of fee payment: 11

Ref country code: IS

Payment date: 20210622

Year of fee payment: 11

REG Reference to a national code

Ref country code: GR

Ref legal event code: NF

Ref document number: 20160402422

Country of ref document: GR

Effective date: 20210915

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: HR

Payment date: 20210701

Year of fee payment: 11

Ref country code: BG

Payment date: 20210701

Year of fee payment: 11

Ref country code: CY

Payment date: 20210623

Year of fee payment: 11

Ref country code: AT

Payment date: 20210624

Year of fee payment: 11

REG Reference to a national code

Ref country code: HR

Ref legal event code: PNEV

Ref document number: P20161352

Country of ref document: HR

REG Reference to a national code

Ref country code: EE

Ref legal event code: MF4A

Ref document number: E012605

Country of ref document: EE

Effective date: 20210907

Ref country code: AT

Ref legal event code: MA03

Ref document number: 820414

Country of ref document: AT

Kind code of ref document: T

Effective date: 20210615

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20210802

Year of fee payment: 11

Ref country code: SI

Payment date: 20210628

Year of fee payment: 11

Ref country code: RS

Payment date: 20210702

Year of fee payment: 11

Ref country code: HU

Payment date: 20210628

Year of fee payment: 11

REG Reference to a national code

Ref country code: SE

Ref legal event code: ECNC

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MT

Payment date: 20210702

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: AL

Payment date: 20210730

Year of fee payment: 11

REG Reference to a national code

Ref country code: SI

Ref legal event code: KO00

Effective date: 20230314